Language selection

Search

Patent 2818308 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2818308
(54) English Title: BIFUNCTIONAL MOLECULES WITH ANTIBODY-RECRUITING AND ENTRY INHIBITORY ACTIVITY AGAINST THE HUMAN IMMUNODEFICIENCY VIRUS
(54) French Title: MOLECULES BIFONCTIONNELLES DOTEES D'UNE ACTIVITE DE RECRUTEMENT D'ANTICORPS ET D'INHIBITION DE L'ENTREE DU VIRUS DIRIGEES CONTRE LE VIRUS DE L'IMMUNODEFICIENCE HUMAINE
Status: Deemed Abandoned and Beyond the Period of Reinstatement - Pending Response to Notice of Disregarded Communication
Bibliographic Data
(51) International Patent Classification (IPC):
  • C7D 405/14 (2006.01)
  • A61K 31/496 (2006.01)
  • A61P 31/18 (2006.01)
(72) Inventors :
  • SPIEGEL, DAVID (United States of America)
  • PARKER, CHRISTOPHER (United States of America)
(73) Owners :
  • YALE UNIVERSITY
(71) Applicants :
  • YALE UNIVERSITY (United States of America)
(74) Agent: WILSON LUE LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2011-11-17
(87) Open to Public Inspection: 2012-05-24
Examination requested: 2016-11-16
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2011/061174
(87) International Publication Number: US2011061174
(85) National Entry: 2013-05-16

(30) Application Priority Data:
Application No. Country/Territory Date
61/414,977 (United States of America) 2010-11-18
61/522,518 (United States of America) 2011-08-11

Abstracts

English Abstract

The present invention is directed to new bifunctional compounds and methods for treating HIV infections. The bifunctional small molecules, generally referred to as ARM- HI's, function through orthogonal pathways, by inhibiting the gpl20-CD4 interaction, and by recruiting anti-DNP antibodies to gpl20-expressing cells, thereby preventing cell infection and spread of HIV. It has been shown that ARM-HI's bind to gpl20 and gp-120 expressing cells competitively with CD4, thereby decreasing viral infectivity as shown by an MT-2 cell assay, the binding leading to formation of a ternary complex by recruiting anti-DNP antibodies to bind thereto, the antibodies present in the ternary complex promoting the complement-dependent destruction of the gpl20-expressing cells. Compounds and methods are described herein.


French Abstract

La présente invention concerne de nouveaux composés bifonctionnels et des procédés de traitement d'infections par le VIH. Les petites molécules bifonctionnelles, généralement désignées sous le nom ARM-HI, fonctionnent par le biais de voies orthogonales, en inhibant l'interaction gpl20-CD4, et en recrutant des anticorps anti-DNP dans des cellules exprimant le gp120, empêchant ainsi l'infection des cellules et la propagation du VIH. Il s'est avéré que les ARM-HI se lient au gp120 et aux cellules exprimant le gp-120 de manière compétitive avec le CD4, diminuant ainsi l'infectiosité virale tel que démontré par un dosage des cellules MT-2, la liaison menant à la formation d'un complexe ternaire grâce au recrutement des anticorps anti-DNP destinés à se lier à celui-ci, les anticorps présents dans le complexe ternaire favorisant la destruction dépendante du complément des cellules exprimant le gp120. L'invention concerne ainsi des composés et des procédés.

Claims

Note: Claims are shown in the official language in which they were submitted.


64
Claims:
1. A compound according to the chemical structure:
<IMG>
where <IMG> is an antibody binding terminus (moiety) comprising a hapten
which is
capable of binding to an antibody present in a patient;
<IMG> is a linker molecule which chemical links ABT to R Y or directly to the
indole
moiety at the carbon atom to which R Y is attached and which optionally
includes a connector
CT which may be a bond or a connector molecule;
<IMG>
is an aromatic or heteroaromatic group;
R Y is absent or is an optionally substituted aryl or heteroaryl group or O,
(CH2)j, NR1, -S-,
-NHC(O)-, -NHC(O)NH-, S(O), S(O)2, -S(O)2O, -OS(O)2, or OS(O)2O;
X2 is H, -(CH2)n OH, -(CH2)n COOH, optionally substituted C1-C6 alkyl, -(CH2)O-
(C1-C6
alkyl), -(CH2)n C(O)-(C1-C6 alkyl), -(CH2)n NHC(O)-R1, -(CH2)n NR1R2, -(CH2)n
C(O)-NR1R2,
-(CH2O)n H, -(CH2O)n COOH, -(OCH2)n O-(C1-C6 alkyl), -(CH2O)n C(O)-(C1-C6
alkyl),
-(OCH2)n NHC(O)-R1, -(CH2O)n C(O)-NR1R2, NO2, CN or halogen;

65
X3 is H, -(CH2)OH, -(CH2)n COOH, optionally substituted C1-C6 alkyl, -(CH2)n O-
(C1-C6
alkyl), -(CH2)n C(O)-(C1-C6 alkyl), -(CH2)n NR1R2,, -(CH2)n NHC(O)-R1, -(CH2)n
C(O)-NR1R2,
-(CH2O)n H, -(CH2O)n COOH, -(OCH2)n O-(C1-C6 alkyl), -(CH2O)n C(O)-(C1-C6
alkyl),
-(OCH2)n NHC(O)-114, -(CH2O)n C(O)-NR1R2, NO2, CN, halogen or a monocyclic
aryl or
heteroaryl group which itself is optionally substituted;
R1 is H or a C1-C3 alkyl group;
R1 and R2 are each independently H or a C1-C6 alkyl group;
i is 0 or 1;
j is 1, 2 or 3;
k is 0,1,2 or 3;
n is 0, 1, 2, 3, 4, 5, 6;
Y3 is H or a C1-C3 alkyl group; and
R N is H or a C1-C3 alkyl group which is optionally substituted with one or
two hydroxyl
groups or up to three halogen groups,
or a pharmaceutically acceptable salt, enantiomer, solvate or polymorph
thereof.
2. The compound according to claim 1 according to the formula:
<IMG>

66
where <IMG> is a monocyclic or bicyclic aryl or heteroaryl group according
to the
chemical structure:
<IMG>
where W is H, -(CH2)n OH, -(CH2)n COOH, C1-C6 alkyl,
-(CH2)n O-(C1-C6 alkyl), -(CH2)n C(O)-(C1-C6 alkyl), -(CH2)n NHC(O)-R1, -
(CH2)n C(O)-
NR1R2, -(CH2O)n OH, -(CH2O)n COOH, C1-C6 alkyl, -(CH2O)n O-(C1-C6 alkyl), -
(CH2O)n C(O)-(C1-C6 alkyl), -(CH2O)n NHC(O)-R1, -(CH2O)n C(O)-NR1R2, NO2, CN,
halogen

67
(F, CI, Br, I, preferably F or CI) or a monocyclic aryl or heteroaryl group
which itself is
optionally substituted (especially an optionally substituted benzoyl or benzyl
group);
W' is H, -(CH2)n OH, -(CH2)n COOH, C1-C6 alkyl, -(CH2)n O-(C1-C6 alkyl) or
halogen
(preferably F or Cl);
<IMG> is a group according to chemical structure:
<IMG>
where W2 is H, -(CH2)n OH, -(CH2)n COOH, C1-C6 alkyl, -(CH2)n O-(C1-C6 alkyl),
-(CH2)n C(O)-(C1-C6 alkyl), -(CH2)n NHC(O)-R1, -(CH2)n C(O)-NR1R2, -(CH2O)n
OH,
-(CH2O)n COOH, C1-C6 alkyl, -(CH2O)n O-(C1-C6 alkyl), -(CH2O)n C(O)-(C1-C6
alkyl),
-(CH2O)n NHC(O)-R1, -(CH2O)n C(O)-NR1R2,NO2, CN or halogen (preferably F or
Cl);
X is a group -(CH2)n NH-, -(CH2)NHC(O)-, -(CH2)n O-, 0(CH2)m-, -(CH2)n S-, -
(CH2)n S(O)-,
-(CH2)n SO2- or -(CH2)n NH-C(O)-NH- which links <IMG> to the linker;
Y is O, S or N-R where R is H or a C1-C3 alkyl group;
X2 is H, -(CH2)n OH, -(CH2)n COOH, C1-C6 alkyl, -(CH2)n O-(C1-C6 alkyl),
-(CH2)n C(O)-(C1-C6 alkyl), -(CH2)n NHC(O)-R1, -(CH2)n C(O)-NR1R2, -(CH2O)n
OH,
-(CH2O)n COOH, C1-C6 alkyl, -(CH2O)n O-(C1-C6 alkyl),

68
-(CH2O)n C(O)-(C1-C6 alkyl), -(CH2O)n NHC(O)-R1 or -(CH2O)n C(O)-NR1R2,NO2;
R1 and R2 are each independently H or a C1-C6 alkyl group;
Y3 is H or a C1-C3 alkyl group (preferably, disposed out of or into the plane,
preferably out of
the plane on the chiral carbon; and
R N is H or a C1-C3 alkyl group which is optionally substituted with one or
two hydroxyl
groups or up to three halogen groups (preferably F);
i is 0 or 1, preferably 1; and
m is 1, 2, 3, 4 or 5, preferably 1, 2 or 3, more preferably 1; and
each n is independently 0, 1, 2 or 3;
<IMG>
is an antibody binding terminus (moiety) comprising a hapten which is capable
of
binding to an antibody present in a patient; and
<IMG> is a linker molecule which links <IMG> to
X; or
a pharmaceutically acceptable salt, enantiomer, solvate or polymorph thereof
3. The compound according to claim 1 according to the formula:

69
<IMG>
where <IMG> is a monocyclic or bicyclic aryl or heteroaryl group according
to the
chemical structure:

70
<IMG>
where W is H, -(CH2)n OH, -(CH2)n COOH, optionally substituted C1-C6 alkyl,
-(CH2)n O-(C1-C6 alkyl), -(CH2)n C(O)-(C1-C6 alkyl), -(CH2)n NR1R2õ -(CH2)n
NHC(O)-R1,
-(CH2)n C(O)-NR1R2, -(CH2O)n H, -(CH2O)n COOH, -(OCH2)n O-(C1-C6 alkyl),
-(CH2O)n C(O)-(C1-C6 alkyl), -(OCH2)n NHC(O)-R1, -(CH2O)n C(O)-NR1R2, NO2, CN,
halogen (F, CI, Br, I, preferably F or Cl) or a monocyclic aryl or heteroaryl
group which itself
is optionally substituted;
W' is H, -(CH2)n OH, -(CH2)n COOH, C1-C6 alkyl, -(CH2)n O-(C1-C6 alkyl) or
halogen;

71
<IMG>
is a group according to chemical structure:
<IMG>
where W2 is H, -(CH2)n OH, -(CH2)n COOH, optionally substituted C1-C6 alkyl, -
(CH2)n O -(C1-
C6 alkyl), -(CH2)n C(O)-(C1-C6 alkyl), -(CH2)n NR1R2, -(CH2)n NHC(O)-R1, -
(CH2)n C(O)-
NR1R2 -(CH2O)n H, -(CH2O)n COOH, -(OCH2O)n-(C1-C6 alkyl), -(CH2O)n C(O)-(C1-C6
alkyl),
-(OCH2)n NHC(O)-R1, -(CH2O)n C(O)-NR1R2, NO2, CN or halogen (preferably F or
Cl);
X is a bond or a group -(CH2)n NH-, -(CH2)n NHC(O)-, -(CH2)n O-, -(CH2)m-, -
(CH2)n S-,
-(CH2)n S(O)-, -(CH2)n SO2- or -(CH2)n NH-C(O)-NH- which links <IMG> to
<IMG>
Y is O, S or N-R where R is H or a C1-C3 alkyl group;
R1 and R2 are each independently H or a C1-C6 alkyl group;
Y3 is H or CH3;
<IMG> is a linker group linking <IMG> through X to
said dinitrophenyl amine
group;

72
m is 1, 2, 3, 4 or 5; and
each n is independently 0, 1, 2, 3, 4, 5 or 6, or
a pharmaceutically acceptable salt, enantiomer, solvate or polymorph thereof.
4. The compound according to claim 1 wherein R Y is an optionally substituted
aryl or
heteroaryl group.
5. The compound according to claim 4 wherein R Y is an optionally substituted
phenyl, naphthyl, pyridyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl,
furanyl, tetrazolyl, or
thiophenyl group.
6. The compound according to any of claims 1 and 3-5 wherein said ARYL group
is
an optionally substituted phenyl group or a quinoline, isoquinoline,
phthalazine or
quinazoline group.
7. The compound according to any of claims 1 or 4-6 wherein said R Y group is
substituted with a -NH-, -NHCO-, -O-, -CH2-, -S- or -NHC(O)NH- group which
links the R Y
group to the linker group.
8. The compound according to any of claims 1 and 4-7 wherein RN is H, X2 is H
or
OCH3; X3 is H, OCH3, CH3 or NO2; Y3 is H or CH3 (racemic or enantiomeric); and
k and i
are each 1, or a pharmaceutically acceptable salt, enantiomer, solvate or
polymorph thereof.
9. The compound according to any of claims 1-7 wherein Y3 is a CH3 group and
said
CH3 group is disposed outward from the plane.
10. The compound according to any of claims 1 and 3-8 wherein R Y is an
optionally
substituted furanyl or tetrazole group.
11. The compound according to claim 2 or 3 wherein [ARYL2] is a

73
<IMG>
group.
12. The compound according to any of claims 1-11 wherein said linker comprises
a
polyethylene glycol having from 1 to 8 glycol units or a group according to
the formula:
<IMG>
where each n is independently 0, 1, 2, 3, 4, 5
or 6.
13. The compound according to claim 12 wherein said polyethylene glycol linker
has
1 to 6 glycol units.
14. The compound according to claim 1 or 2 wherein in said
<IMG>
where each n is independently 0, 1, 2, 3, 4, 5 or 6 or [LINKER] is a
polyethylene glycol
linker having from 1 to 8 glycol units and ABT is
<IMG>
15. The compound according to claim 2 or 3 wherein said [ARYL1] group is
selected
from the group consisting of phenyl, nitrophenyl, o-, m-, or p-toluyl, o-, m-
or p-ethylphenyl,
o-, m- or p-isopropylphenyl, naphthyl, quinidinyl, o-,m- or p-phenol, 3,5-
dihydroxylphenyl,
o-, m- or p-hydroxymethylphenyl or a 2-, 3-, or 4-pyridyl group.

74
16. The compound according to any of claims 1 or 4-10 wherein said [ARYL]
group
is selected from the group consisting of phenyl, nitrophenyl, o-, m-, or p-
toluyl, o-, m- or p-
ethylphenyl, o-, m- or p-isopropylphenyl, naphthyl, quinidinyl, o-,m- or p-
phenol, 3,5-
dihydroxylphenyl, o-, m- or p-hydroxymethylphenyl or a 2-, 3-, or 4-pyridyl
group.
17. The compound according to any of claims 1 or 4-10 wherein R Y is a phenyl
group
substituted with a C1-C6 alkyl, hydroxyl or methylalcohol, an optionally
substituted furanyl
group or tetrazolyl group.
18. The compound according to any of claims 1-2, 4-10 and 12-13 wherein ABT
comprises a moiety according to the chemical formula:
<IMG>

75
<IMG>
where Y' is H or NO2;
X is O, CH2, NR1, S(O), S(O)2, -S(O)2O, -OS(O)2, or OS(O)2O;
R1 is H, a C1-C3 alkyl group, or a -C(O)(C1-C3) group;
Xs is OH or NHAc;.
X R is O or S;
X M is O or S;
X' is CH2, O, N-R1, or S;
R1' is H or C1-C3 alkyl;
Z is a bond, a monosaccharide, disaccharide, oligosaccharide, glycoprotein or
glycolipid;
Xs is OH or NHAc;
X b is a bond, O, CH2, NR1or S;
X" is O, CH2, NR1; and
R1 is H, a C1-C3 alkyl group or a -C(O)(C1-C3) group.
19. The compound according to any of claims 1-2, 4-10, 12-13 or 16-17 wherein
said ABT moiety comprises a dinitrophenyl group.
20. The compound according to claim 18 wherein X' is O or N-R1' and R1' is H.
21. The compound according to claim 18 or 20 wherein X' is O.

76
22. The compound according to any of claims 1 and 3-9, 11-12 or 15 wherein
said
ABT moiety comprises a group according to the chemical formula:
<IMG>
where Y' is H or NO2; and
X is O, CH2, Nil', S(O), S(O)2, -S(O)2O, -OS(O)2, or OS(O)2O.
23. The compound according to any of claims 1-13 wherein said linker is
<IMG>
where R a is H, C1-C3 alkyl or alkanol or forms a cyclic ring with R3 and R3
is a side chain
derived from an amino acid; and m is an integer from 1 to 15.
24. The compound according to any of claims 1-13, 15 or 19 wherein said linker
is
<IMG>
and n is 1, 2, 3, 4, 5, or 6.
25. The compound according to any of claims 1-10 or 12-13 wherein said linker
is an
amino acid linker or a polypropylene glycol or polypropylene-co-polyethylene
glycol linker
having between 1 and 15 glycol units.

77
26. The compound according to any of claims 1-13 wherein said linker is a
compound
according to the chemical formula:
<IMG>
where Z and Z' are each independently a bond, -(CH2)i-O, -(CH2)-S, -(CH2)i-N-
R,
<IMG>
wherein said -(CH2), group, if present in Z or Z', is bonded to a connector,
ABT or CBT;
Each R is H, or a C1-C3 alkyl or alkanol group;
Each R2 is independently H or a C1-C3 alkyl group;
Each Y is independently a bond, O, S or N-R;
Each i is independently 1 to 50;
D is
<IMG>
a bond, with the proviso that Z, Z' and D are not each simultaneously bonds;
j is 1 to 100;
m' is 1 to 100;
n is 1 to 100;
X1 is O, S or N-R; and
R is as described above, or a pharmaceutical salt thereof

78
or a pharmaceutically acceptable salt, solvate or polymorph thereof.
27. The compound according to any of claims 1-2, 4-10 and 12-13 wherein
said
ABT moiety is selected from the group consisting of the moieties having the
following
structures:
<IMG>
wherein said DNP group is optionally linked through an X group to said amino
acid moiety,
where X is CH2, S(O), S(O)2, -S(O)2O, -OS(O)2, or OS(O)2O.
28. The compound according to claim 27 wherein said ABT group is

79
<IMG>
29. The compound according to claim 1, 2 or 3 which is
<IMG>
30. The compound according to claim 2 wherein X2 is H or OCH3;
<IMG>
W is H or NO2;

80
<IMG>
Y is O;
X is a group ¨(CH2n NH-, -(CH2)n NHC(O)-, -(CH2)n O-, -(CH2)m-, -(CH2)n S-, -
(CH2)n S(O)-, -
(CH2)n SO2- or ¨(CH2)n NH-C(O)-NH-;
R N is H;
Y3 is H or a CH3 group disposed outward from the plane;
<IMG>
ABT is
<IMG>
is a linking group comprising a polyethylene glycol having from 1 to 8 glycol
units;
i is 1;
m is 1, 2, 3, 4 or 5;
each n is independently 0, 1, 2, 3, 4, 5 or 6, or
a pharmaceutically acceptable salt, enantiomer, solvate or polymorph thereof.
31. A pharmaceutical composition comprising an effective amount of a
bifunctional
compound according to any of claims 1-30 combination with a pharmaceutically
acceptable
carrier, additive or excipient.
32. The composition according to claim 31 wherein said composition further
comprises an effective amount of an additional anti-HIV agent.
33. The composition according to claim 32 wherein said additional anti-HIV
agent is
selected from the group consisting of nucleoside reverse transcriptase
inhibitors (NRTI), non-
nucloeoside reverse transcriptase inhibitors, protease inhibitors and fusion
inhibitors.

81
34. The composition according to claim 32 wherein said additional anti-HIV
agent is
selected from the group consisting of Amprenivir, Abacavir, Acemannan,
Acyclovir, AD-
439, AD-519, Adefovir dipivoxil, Alpha Interferon, Ansamycin, 097, AR 177,
Beta-fluoro-
ddA, BMS-232623 (CGP-73547), BMS-234475 (CGP-61755), CI-1012, Cidofovir,
Curdlan
sulfate, Cytomegalovirus Immune globin, Ganciclovir, Dideoxyinosine, DMP-450,
Efavirenz
(DMP-266), ELIO, Famciclovir, FTC, GS 840, HBY097, Hypericin, Recombinant
Human
Interferon Beta, Interferon alfa-n3, Indinavir, ISIS-2922, KNI-272, Lamivudine
(3TC),
Lobucavir, Nelfinavir, Nevirapine, Novapren, Peptide T Octapeptide Sequence,
Trisodium
Phosphonoformate, PNU-140690, Probucol, RBC-CD4, Ritonavir, Saquinavir,
Valaciclovir,
Virazole Ribavirin, VX-478, Zalcitabine, Zidovudine (AZT), Tenofovir
diisoproxil fumarate
salt, Combivir, Abacavir succinate, T-20), AS-101, Bropirimine, CL246, EL10,
FP-21399,
Gamma Interferon, Granulocyte Macrophage Colony Stimulating Factor (GM-CSF),
HIV
Core Particle Immunostimulant, Interleukin-2 (IL-2), Immune Globulin
Intravenous,
IMREG-1, IMREG-2, Imuthiol Diethyl Dithio Carbamate, Alpha-2 Interferon,
Methionine-
Enkephalin, MTP-PE (Muramyl-Tripeptide), Granulocyte Colony Stimulating Factor
(GCSF), Remune, rCD4 (Recombinant Soluble Human CD4-IgG), rCD4-IgG Hybrids,
Recombinant Soluble Human CD4, Interferon Alfa 2a, SK&F1-6528, Soluble T4,
Thymopentin, Tumor Necrosis Factor (TNF), AK602, Alovudine, Amdoxovir, AMD070,
A tazanavir (Reyataz), AVX754 (apricitabine), Bevirimat, BI-201, BMS-378806,
BMS-
488043, BMS-707035, C31G, Carbopol 974P, Calanolide A, Carrageenan, Cellulose
sulfate,
Cyanovirin-N, Darunavir, Delavirdine, Didanosine (Videx), Efavirenz,
Elvucitabine,
Emtricitabine, Fosamprenavir (Lexiva), Fozivudine tidoxil, GS 9137, GSK-
873,140
(aplaviroc), GSK- 364735, GW640385 (brecanavir), HG0004, HGTV43, INCB9471, KP-
1461, Lopinavir, Mifepristone (VGX410), MK-0518, PPL-100, PRO 140, PRO 542,
PRO
2000, Racivir, SCH-D (vicriviroc), SPO1A, SPL7013, TAK-652, Tipranavir
(Aptivus), TNX-
355, TMC125 (etravirine), UC-781, UK-427,857 (Maraviroc), Valproic acid,
VRX496,
Zalcitabine, Valganciclovir, Clindamycin with Primaquine, Fluconazole
Pastille, Nystatin
Pastille, Eflornithine, Pentamidine, Isethionate, Trimethoprim,
Trimethoprim/sulfa,
Piritrexim, Pentamidine isethionate, Spiramycin, Intraconazole-R51211,
Trimetrexate,
Daunorubicin, Recombinant Human Erythropoietin, Recombinant Human Growth
Hormone,
Megestrol Acetate, Testosterone, Aldesleukin (Proleukin), Amphotericin B,
Azithromycin
(Zithromax), Calcium hydroxyapatite, Doxorubicin, Dronabinol, Entecavir,
Epoetin alfa,

82
Etoposide, Fluconazole, Isoniazid, Itraconazole (Sporanox), Megestrol,
Paclitaxel (Taxol),
Peginterferon alfa-2, Poly-L-lactic acid (Sculptra), Rifabutin (Mycobutin),
Rifampin,
Somatropin and Sulfamethoxazole/Trimethoprim.
35. The composition according to claim 32 wherein said additional anti-HIV
agent is
selected from the group consisting of 3TC (Lamivudine), AZT (Zidovudine),
(+FTC, ddI
(Didanosine), ddC (zalcitabine), abacavir (ABC), tenofovir (PMPA), D-D4FC
(Reverset),
D4T (Stavudine), Racivir, L-FddC, L-FD4C, NVP (Nevirapine), DLV (Delavirdine),
EFV
(Efavirenz), SQVM (Saquinavir mesylate), RTV (Ritonavir), IDV (Indinavir), SQV
(Saquinavir), NFV (Nelfmavir), APV (Amprenavir), LPV (Lopinavir), T20, fuseon
and
mixtures thereof.
36. The composition according to any of claims 31-35 in oral dosage form.
37. The composition according to any of claims 31-35 in parenteral dosage
form.
38. The composition according to claim 31-35 in topical dosage form.
39. The composition according to claim 37 wherein said parenteral dosage form
is an
intravenous dosage form.
40. Use of a compound according to any of claims 1-30 in the manufacture of a
medicament for the treatment of an HIV infection in a patient.
41. Use of a compound according to any of claims 1-30 in the manufacture of a
medicament for reducing the likelihood that a patient at risk for an HIV
infection will
contract an HIV infection.
42. Use of a compound according to any of claims 1-30 in the manufacture of a
medicament for reducing the likelihood of AIDS or ARC in a patient infected
with HIV.

83
43. Use of a compound according to any of claims 1-30 in the manufacture of a
medicament for reducing or abolishing HIV infected CD cells in an HIV infected
patient.
44. Use of a compound according to any of claims 1-30 in the manufacture of a
medament for inhibiting or abolishing HIV in a patient in need.

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02818308 2013-05-16
Bifunctional Molecules with Antibody-Recruiting and Entry Inhibitory Activity
Against
the Human Immunodeficiency Virus
Field of the Invention
The present invention relates to bifunctional molecules for inhibiting Human
Immunodeficiency Virus (HIV) infection through binding to the HIV glycoprotein
gp 120,
while also engaging in antibody-recruiting for attracting and binding
antibodies which
combat the bound HIV.
Priority Claim and Grant Support
This application claims priority from provisional application serial numbers
US61/414,977 entitled, Development of Small Molecule Antibody Recruiting
Therapeutics
for the Treatment of HIV, ARM-H113 Species and Synthetic Route, filed November
18,
2010, and US61/522,518, filed August 11, 2011, entitled, Bifunctional
Molecules with
Antibody-Recruiting and Entry Inhibitory Activity Against the Human
Immunodeficiency
Virus, the entire contents of said applications being incorporated by
reference herein.
This invention was made with support from the National Institutes of Health
grant no.
DP220D002913. The government retains rights in the invention.
Background and Discussion of the Invention
In recent years, antibody based therapeutics have become important instruments
in
treating human disease. (Brekke, 0. H.; Sandlie. I. Nat. Rev. Drug Discovery
2003,2, 52-62.)
Current antibody-based therapeutics function either by blocking the effector
actions of
pathological molecules, or by targeting specific epitopes on cell surfaces for
immune-
mediated destruction. However, these approaches suffer from certain
limitations, including
severe side effects, lack of oral bioavailability, and high cost. (Allen. T.
M. Nat. Rev. Cancer
2002,2, 750-763.) Thus, alternative methods are being sought that would still
exploit the
powerful cytolytic potential of antibodies already present in the human blood
stream yet
avoid many of these disadvantages.

CA 02818308 2013-05-16
2
Acquired Immune Deficiency Syndrome (AIDS) is a world-wide epidemic that has
claimed many lives and severely debilitated many more through immune
suppression. A
great deal of effort has been directed toward the development of a vaccine for
this disease,
without success. The failure to develop an effective vaccine underscores the
need for new
prevention and treatment strategies toward this disease.
The human immune system is highly versatile in its ability to target and
destroy
foreign pathogens. HIV, however, is an elusive virus to the body's immune
system which
has evolved mechanisms both to evade and to destroy the immune response of
human hosts
in the process of causing AIDS. Researchers recently found that one of the
shortfalls of the
human immune system in combating HIV is also one the antibody's stronger point
against
other viruses; its structure.
An antibody is a Y-shaped molecule with two epitopes comprising antigen
recognizing proteins on the two Y tips. These two epitopes allow the antibody
to bind to two
proteins on the antigen's surface, creating a stronger bond when compared to a
one-epitope
protein bond. Viruses have proteins extending from their viral coat, which are
the proteins
the antibodies bind to.
HIV has fewer proteins than normal viruses. The proteins are placed farther
apart and
this structural difference is believed responsible for the antibody's epitopes
being unable to
bind to two different HIV surface proteins (See Klein et al. "Examination of
the contributions
of size and avidity to the neutralization mechanisms of the anti-HIV
antibodies b12 and
4E10" Proceedings of the National Academy of Sciences, 2009 Abstract).
HIV has also been shown to bind to a surface molecule known as the CD4 or T4
receptor, which is present on various cells susceptible to HIV infection,
including T
lymphocytes and macrophages. (See Shaw et al., Science 226, pp. 1165-1171 for
a discussion
of tropism of HTLV-III.)
A few methods for recruiting naturally occurring antibodies to cancer cells
have
appeared in the literature, but none are believed to have been explored
directed to HIV (See
Carlson, C.; Mowery, P.; Owen, R.; Dykhuizen, E. C.: Kiessling, L. ACS Chem.
Biol. 2007,
2, 119-127; Owen. R.; Carlson, C; Xu, J.; Mowery, P.; Fasella, E.; Kiessling,
L.

CA 02818308 2013-05-16
3
ChemBioChem 2007, 8, 68-82; Popkov. M.: Gonzalez, E.; Sinha, S.; Barbas, C.
Proc. Natl.
Acad. Sci. U.S.A. 2009, 106,4378-4383; Popkov, M.; Rader. C; Gonzalez, B.;
Sinha. S.;
Barbas, C. Intl. J. Cancer 2006, 119, 1194-1207; (25) Low, P.; Herne, W.;
Doorneweerd, D.
Acc. Chem. Res. 2008,41, 120-129; Lu, Y.: You, F.; Vlahov, I.; Westrick. E.;
Fan, M.; Low,
P. S.; Leamon, C. P. Mol. Pharm. 2007,4, 695-706.
(27) Rader. C.; Sinha, S. C.; Popkov, M.; Lerner, R. A.; Barnas, C. F. Proc.
Natl. Acad. Sci.
U.S.A. 2003, 100, 5396-5400), bacteria (Bertozzi, C. R.; Bednarski, M. D. J.
Am. Chem. Soc.
1992, 114, 5543-5546; Bertozzi, C. R.; Bednarski. M. D. J. Am. Chem. Soc.
1992. 114,
2242-2245; Li, J.: Zacharek, S.; Chen, X.; Wang, J. Q.: Zhang, W.; Janczuk,
A.; Wang, P. G.
Bioorg. Med. Chem. 1999, 7, 1549-1558; Krishnamurthy, V. M.; Quinton, L. J.;
Estroff. L.
A.; Metallo, S . J.; Isaacs, J. M.; Mizgerd, J. P; Whitesides, G. M.
Biomaterials 2006, 27,
3663-3674), and viruses ((32) Shokat, K. M.; Schultz, P. G. J. Am. Chem. Soc.
1991, 113,
1861-1862; Naicker, K. P.: Li, H.; Heredia, A.; Song, H.; Wang, L. Org.
Biomol. Chem.
2004,2, 660-664; Perdomo, M. F.: Levi. M.; Ilberg, M. S.; Vahlne, A. Proc.
Natl. Acad. Sci.
U.S.A. 2008, 105,6).
In the HIV realm, most approaches have relied upon protein-or peptide-based
antibody targeting constructs. For example, Shokat and Schultz (Shokat, K. M.;
Schultz, P.
G. J. Am. Chem. Soc. 1991, 113, 1861-1862) first demonstrated that anti-DNP
antibodies
could be redirected to immobilized protein targets (gp120 and streptavidin) as
a therapeutic
strategy toward HIV. More recent work in this vein has employed peptide-R-Gal
conjugates
to target human anti-Gal antibodies to HIV-infected cells. (Naicker, K. P.:
Li, H.; Heredia,
A.; Song, H.; Wang, L. Org. Mom& Chem. 2004,2, 660-664; Perdomo, M. F.: Levi.
M.;
Ilberg, M. S.; Vahlne, A. Proc. Natl. Acad. Sci. U.S.A. 2008, 105,6)
While these peptide conjugates were shown to be effective in killing Env-
expressing
cells, they were also found to exhibit some non specific cytotoxicity.
Bertozzi, C. R.;
Bednarski, M. D. J. Am. Chem. Soc. 1992, 114, 5543-5546.
The present work sought to address these deficiencies, by providing
compositions for
treating HIV infection which can improve the immune system's ability to
respond to HIV
infection. We have discovered one way to assist the body is to recruit
existing antibodies to
attack HIV. Specifically, we have developed bifunctional molecules (Corson. T.
W.; Aberle,
N.; Crews, C. M. ACS Chem. Biol. 2008, 3, 677-692) capable of which inhibit
the

CA 02818308 2013-05-16
4
pathogenic behavior of HIV through two distinct mechanisms: (1) by interfering
with viral
entry via antagonism of the interaction between the viral envelope protein
gp120 and the
human protein CD4, and (2) by recruiting anti-dinitrophenyl ("anti-DNP")
antibodies, a
population of antibodies present in high concentrations in the human
bloodstream, to the
surface of the HIV virus and/or HIV-infected cells.
Antibodies recognizing the DNP epitope have been estimated to constitute 1 %
of
circulating IgM and 0.8% of circulating IgG. See: (a) Karjalainen, K., Makela.
0. Eur. J.
Immunol. 1976, 6, 88-93. (b) Farah, F. S. Immunology 1973,25, 217-226. The
prevalence of
anti-DNP antibodies has been estimated at between 18 and 90% of humans. See:
(c) Ortega,
E.; Kostovetzky. M.; Larralde, C. Mol. Immunol. 1984,21, 883-888. (d)
Jormalainen, S.;
Makela. 0. Eur. J. Immunol. 1971, 1,471-478. Consequently, administration of a
bifunctional molecule which can recruit these existing antibodies to attack
HIV in a patient
suffering from HIV infection may provide a basis for an effective treatment
for the symptoms
associated with HIV infection.
Summary Of The Invention
The present invention relates to ARM-HII ("Antibody-Recruiting Molecules
targeting
HIV Improved") compounds according to the general formula:
0 ARYL
X3)k
X2 0
i y3
\ RN
RY
AB

CA 02818308 2013-05-16
ABT
Where _____ is an antibody binding terminus (moiety) comprising a hapten
which is
capable of binding to an antibody present in a patient (preferably a DNP
group);
LINKER
_______ is a linker molecule which chemical links ABT to RY or directly to the
indole
moiety at the carbon atom to which RI( is attached and which optionally
includes a connector
CT which may be a bond or a connector molecule;
ARYL
is a an aromatic or heteroaromatic group, preferably a monocyclic or bicyclic
aromatic or heteroaromatic group;
121( is absent or is an optionally substituted aryl or heteroaryl group or 0,
(CH2)i, NR1, -S-,
-NHC(0)-, -NHC(0)NH-, S(0), S(0)2, -S(0)20, -OS(0)2, or OS(0)20 (the aryl or
heteroaryl
group also may be substituted with these and/or other groups and/or each of
these groups also
may link the aryl or heteroaryl group to the indole moiety);
X2 is H, -(CH2)õOH, -(CH2).COOH, C1-C6 alkyl, -(CH2).0-(C1-C6 alkyl),
-(CH2)6C(0)-(CI-C6 alkyl), -(CH2)NHC(0)-R1, -(CH2).C(0)-NRIR2, -(CH20)nH,
-(CH20)11C00il, -(OCH2)n0-(C1-C6 alkyl), -(CH20)nC(0)-(C1-C6 alkyl), -
(OCH2)NHC(0)-
RI, -(CH20)nC(0)-NRIR2, NO2, CN or halogen (F, Cl, Br, I, preferably F or Cl);
X3 is H, -(CH2)60H, -(CH2)COOH, Ci-C6 alkyl, -(CH2)60-(C1-C6 alkyl),
-(CH2)nC(0)-(C1-C6 alkyl), -(CH2)6NHC(0)-R1, -(CH2)6C(0)-NRIR2, -(Cf120)nH,
-(CH20)nCOOil, -(0CH2)110-(C1-C6 alkyl), -(CH20)6C(0)-(C1-C6 alkyl),
-(0CH2)6NHC(0)-R1, -(CH20)õC(0)-NRIR2, NO2, CN, halogen (F, Cl, Br, I,
preferably F or
Cl) or a monocyclic aryl or heteroaryl group which itself is optionally
substituted;
RI is H or a C1-C3 alkyl group;
R1 and R2 are each independently H or a C1-C6 alkyl group;
i is 0 or 1, preferably 1;
j is 1, 2 or 3;
k is 0, 1, 2 or 3, preferably 0, 1 or 2;
n is 0, 1, 2, 3, 4, 5, 6, preferably 0-3;
Y3 is H or a C1-C3 alkyl group (preferably, disposed out of or into the plane,
preferably out of
the plane on the chiral carbon; and
RN is H or a C1-C3 alkyl group which is optionally substituted with one or two
hydroxyl
groups or up to three halogen groups (preferably F)
or a pharmaceutically acceptable salt, enantiomer, solvate or polymorph
thereof.

CA 02818308 2013-05-16
6
In other preferred aspects of the invention, compounds according to the
present
invention may be represented by the chemical structure:
ci\7 ARYL1
x2 0
1 y3
\ 0
RN
ARYL2
NO2
N
N
NI
NO2
ARYLI
Where is a monocyclic or bicyclic aryl or heteroaryl group
according to
the chemical structure:

CA 02818308 2013-05-16
7
N\, ,
f.%-\ ,------'. '-'----.-:-.,,,r-NX--- ----- --- --- =
.---. .. '''''....- - - ' -
N \r 1
X. NV\'
1 I
NN AN ,IXN N
W W 'W
/ /
- .------%\-- ---"---- - -- ''.---'-'''./i = .,---
-- - - v.-. V, ...:\::: S,
L V I I
--; ,.,=:,,..,.--"-'-:----..,.õ----' ,,,-- ------..I '''' -...,,-
''''`.õ/ , ''',,,,,. y õ,.--=---7---,---/..z.õ,õ../ ,
W ' W WI
<Y.----\:
\_________
Y -------, , X:. , N ------- ,
W W W
.Pfj4" .rs\fjj.- .34-14" J-Prj"
NC
w\V c
----==i
W w , \ --,-------./ W' N 17 W' -- .-./ \-
X ---1-2:¨W.
, W W
, ,
' W
sfrsj.-\ W
N
r----CMN ¨W )-:----------\----- , N 1 ¨ I ----- \N¨w *SIV)---N\
N---
N ...õ---..-......_ / N --,--- / N -..._---..._ /
N N , ---- N N -....,- /
, N .
,
Where W is H, -(CH2)n0H, -(CH2)nCOOH, Ci-C6 alkyl,
-(CH2)n0-(C1-C6 alkyl), -(CH2)nC(0)-(C1-C6 alkyl), -(CH2)nNHC(0)-R1,
-(CH2)õC(0)-NR1R2, -(CH20)nH, -(CH20)nCOOH, C1-C6 alkyl, -(CH20)n0-(C1-C6
alkyl),
-(CH20)nC(0)-(C1-C6 alkyl), -(CH20)nNHC(0)-R1, -(CH20).C(0)-NRIR2, NO2, CN,
halogen (F, Cl, Br, I, preferably F or Cl) or a monocyclic aryl or heteroaryl
group which itself
is optionally substituted (especially an optionally substituted benzoyl or
benzyl group);
W' is H, -(CH2).0H, -(CH2)11COOH, C1-C6 alkyl, -(CH2)n0-(C1-C6 alkyl) or
halogen
(preferably F or Cl);
ARYL2 1----x---
(' is a group according to chemical structure:

CA 02818308 2013-05-16
8
/
x\ / x\ /11^ xvirr
)(/,
N ' w2 w2 N ' w2 w2 N
J
\c,f,p= )5,3s- \esssr,
NNY
X
I\ 2
W2
X xx, X
"
X
./=`=rV X w2
r\\
X N¨,c NI_ )( 'ssrrN
N¨X
.=====/
N
Where W2 is H, -(CH2).0H, -(CH2).COOH, Ci-C. alkyl, -(CH2).0-(C1-C6 alkyl),
-(CH2)6C(0)-(Ci-C6 alkyl), -(CH2).NHC(0)-R1, -(CH2).C(0)-NR1R2, -(CH20)nH,
-(CH20)nCOOH, Ci-C. alkyl, -(CH20).04C1-C6 alkyl), -(CH20).C(0)-(CI-C6 alkyl),
-(CH20).NHC(0)-R1, -(CH20).C(0)-NR1R2, NO2, CN or halogen (preferably F or
Cl);
X is a a bond or a group ¨(CH2).NH-, -(CH2).NHC(0)-, -(CH2).0-, -(CH2)m-, -
(CH2).8-,
ARYL2
-(CH2).S(0)-, -(CH2). SO2- or ¨(CH2).NH-C(0)-NH- which links to the
linker;
Y is 0, S or N-R where R is H or a C1-C3 alkyl group;
X2 is H, -(CH2).0H, -(CH2).COOH, Ci-C. alkyl, -(CH2).0-(C1-C6 alkyl),
-(CH2).C(0)-(C1-C6 -(CH2).NHC(0)-R1, -(CH2).C(0)-NRIR2, -(CH20)nH,
-(CH20)nCOOH, C1-C6 alkyl, 40CH2).0-(Ci-C6 alkyl),
-(CH20).C(0)-(Ci-C6 alkyl), -(OCH2).NHC(0)-R1 or -(CH20).C(0)-NR1R2;
RI and R2 are each independently H or a C1-C6 alkyl group;
Y3 is H or a Ci-C3 alkyl group (preferably, disposed out of or into the plane,
preferably out of
the plane on the chiral carbon; and
RN is H or a Ci-C3 alkyl group which is optionally substituted with one or two
hydroxyl
groups or up to three halogen groups (preferably F);
i is 0 or 1, preferably 1;
m is 1, 2, 3, 4 or 5, preferably 1, 2 or 3, more preferably 1; and

CA 02818308 2013-05-16
9
Each n is independently 0, 1, 2, 3,4, 5 or 6, preferably 0, 1, 2 or 3, or
a pharmaceutically acceptable salt, enantiomer, solvate or polymorph thereof.
Preferred bifunctional compounds for use in the present invention include
those where
RY is an optionally substituted aryl or heteroaryl group as otherwise
described herein,
preferably an optionally substituted phenyl (especially a C1-C6 alkyl,
hydroxyl,
methylalcohol), naphthyl, pyridyl (2-, 3- or 4-pyridyl group), thiazolyl (2-,
4- or 5-thiazole),
isothiazolyl, oxazolyl (2-, 4- or 5-oxazole), isoxazolyl, furanyl (2- or 3-
furan) or thiophenyl
(2- or 3-thiophene). The RY group is preferably substituted with a ¨NH-, -NHCO-
, -0-,
-CH2-, -S- or -NHC(0)NH- group which links the RY group (preferably as an aryl
or
heteroaryl group) to the linker group. RN is preferably H, X2 is preferably H
or OCH3; X3 is
preferably H, OCH3 or CH3; Y3 is H or CH3 (racemic or enantiomeric); i is
preferably 1, or a
pharmaceutically acceptable salt, enantiomer, solvate or polymorph thereof.
Preferred Aryll
groups include the groups which appear in the Table 1, hereinbelow and include
phenyl, o-,
m-, or p-toluyl, o-, m- or p-ethylphenyl, o-, m- or p-isopropylphenyl,
naphthyl (preferably 1-
or 2-), o-,m- or p-phenol, 3,5-dihydroxylphenyl, o-, m- or p-
hydroxymethylphenyl or a 2-, 3-,
or 4-pyridyl group.
+
LINKER __________________________ ABT
Preferred groups for use in compounds according
to the present invention may be represented by the chemical structure (which
includes a
connector triazole molecule:
NO2
Nr.(
01 I
NO2
Where each n is independently 0, 1, 2, 3, 4, 5 or 6, preferably 0, 1,2 or 3.
Other linker and ABT groups which may be used are described in the
specification
above and in the examples.

CA 02818308 2013-05-16
In an additional aspect of the invention, a pharmaceutical composition
comprises an
effective amount of a bifunctional compound as described above, optionally and
preferably in
combination with a pharmaceutically acceptable carrier, additive or excipient.
In alternative
aspects, pharmaceutical combination compositions comprise an effective amount
of a
bifunctional compound as described herein, in combination with at least one
additional agent
which is used to treat HIV.
In a further aspect of the invention, compounds according to the present
invention are
used to treat and/or reduce the likelihood of an HIV infection or a secondary
effect of HIV
(such as AIDS, ARC and related disease states or conditions which occur
secondary to an
HIV infection) in a patient. The method of treating and/or reducing the
likelihood of an HIV
infection or secondary effect of an HIV cancer comprises administering to a
patient in need
an effective amount of a bifunctional compound as otherwise described herein
in combination
with a pharmaceutically acceptable carrier, additive or excipient, optionally
in further
combination with at least one additional agent which is effective in treating
and/or reducing
the likelihood of an HIV infection, or one or more of its secondary conditions
or effects.
The present invention also relates to instances in which destruction of CD4
cells
which are infected with HIV (HIV +CD4 cells) may be useful to inhibit latent
HIV infections
from becoming active. In this aspect of the invention, destruction of HIV +CD4
cells in an
HIV positive patient may be used to inhibit or more completely eradicate an
HIV infection
and/or reduce the likelihood of an occurrence or recurrence of HIV in a
patient who is HIV
positive.
The present invention also relates to a method for binding and eliminating HIV
in a
patient comprising administering to a patient infected with HIV, an effective
amount of a
bifunctional compound as otherwise described herein.
Thus, the present invention presents unique, non-peptidic, bifunctional
molecules
which can operate through the bifunctional mechanisms specified above in
treating HIV.
The realization that viruses may exert cell and tissue tropism by attachment
at highly
specific sites on cell membrane receptors has encouraged investigators in the
past to seek

CA 02818308 2013-05-16
11
agents which would bind at the viral receptor sites of cell membranes and thus
prevent
binding of a specific virus to these cells.
Specifically, HIV has been shown to bind to a surface molecule known as the
CD4 or
T4 receptor which is present on various cells susceptible to HIV infection,
including T
lymphocytes and macrophages. The binding occurs via the HIV envelope protein,
gp120.
It is an object of the present invention to provide bifunctional compounds
that would
act to alleviate the symptoms of AIDS by binding a bifunctional molecule which
has a first
terminus for binding to the gp120 envelope protein, the bifunctional molecule
having a
second antibody recruiting terminus which attracts antibodies already
circulating throughout
the body, to form a ternary complex between anti-DNP antibodies and gp120
and/or gp120-
expressing cells, the antibodies attacking the HIV engaged by the bifunctional
molecule.
These bifunctional (which term also includes multifunctional) molecules are
thus generically
referred to herein as "Antibody-Recruiting Molecules targeting HIV Improved"
or "ARM-
HII".
The inventive ARM-HII molecules are "bifunctional" in that they possess a at
least
one pathogen binding terminus (PBT) and at least one antibody recruiting
terminus (ABT)
connected by at least one linker and a connector molecule. The PBT is designed
to bind to
the HIV glycoprotein gp120 (gp120 on the viral membrane as well as gp120
displayed on
infected cells). The ABT is designed to bind and/or recruit antibodies to the
site of the
binding of the bifunctional compound according to the present invention.
In one embodiment of the invention, a bifunctional ARM-HI molecule is
described
which is capable of redirecting a population of anti-hapten (e.g. anti-
dinitrophenyl or anti-
DNP) antibodies, which represent a population of antibodies present in high
concentrations in
the human blood stream ("endogenous antibodies"), to the HIV gp120 Env gene
product.
The Env glycoprotein, a complex between gp120 and membrane-bound gp 41, is
expressed
on both the surface of the HIV virus and on virus-infected cells, especially
CD4 cells.
(Miranda. L. R.; Schaefer, B. C:.; Kupfer. A.; Hu, Z. X.; Franzusoff, A. Proc.
Natl. Acad. Sci.
U.S.A, 2002,99, 8031-8036). The gp120 component of Env mediates the first step
in viral
entry into human cells by binding the protein CD4.

CA 02818308 2013-05-16
12
According to the present invention, a ternary complex is formed between anti-
hapten
(e.g. DNP or other haptent) antibodies, ARM-HI, and Env-expressing cells which
mediates
the complement-dependent destruction of these cells. Further, since ARM-HI
binds gp120
competitively with CD4, it also inhibits the entry of live HIV into human T-
cells. Thus,
ARM-HI has the potential to interfere with the survival of HIV through
multiple
complementary mechanisms, and may also function as a prophylactic.
The ARM-HII compounds of the invention are unique in that they represent a
molecule-based, not a peptide and/or protein based, anti-HIV strategy for
targeting the virus
life cycle through mutually reinforcing molecular mechanisms, inhibiting virus
entry while
targeting Env-expressing cells for immune recognition and clearance. In
general, the ARM-
HII molecules have certain advantages over proteins from a therapeutic
standpoint because of
their low propensity for immunogenicity, high metabolic stability, ready large-
scale
production, and relatively low cost. Molecule based antibody-recruiting
therapeutics such as
ARM-HII have additional benefits over available treatment approaches to HIV.
For example,
directing HIV-infected cells and virus particles to Fcy receptors on antigen-
presenting cells
enhances the presentation of viral antigens on MHC proteins and contributes to
long-lasting
anti-HIV immunity. (See Lu, Y.: You, F.; Vlahov, I.; Westrick. E.; Fan, M.;
Low, P. S.;
Leamon, C. P. Mol. Pharm. 2007, 4, 695-706, Rawool, D. B.; Bitsaktsis, C.; Li,
Y.; Gosselin,
D. R., Lin, Y.; Kurkure, N. Y.; Metzger, D. W.; Gosselin, E. J. J. Immunol,
2008, 180, 5548-
5557) Critically, no non specific cytotoxicity was observed in either MT-2 or
CHO cell lines
in response to the inventive ARM-HI molecules, limiting the possibility of
encountering
serious side effects from treatment therewith.
Furthermore, because anti-hapten (anti-DNP) antibodies are already present in
the
human blood stream, no pre-vaccination is necessary for ARM-HI activity. Also,
the binding
of bifunctional molecule targeting agents to antibodies should prolong their
plasma half-life,
thus increasing their effectiveness. (See Rader. C.; Sinha, S. C.; Popkov, M.;
Lerner, R. A.;
Barbas, C. F. Proc. Natl. Acad. Sci. U.S.A. 2003, 100, 5396-5400)
Elucidation of the molecular details governing the interactions among ARM-HI,
gp120, and anti-DNP antibodies assists in optimization efforts as well as in
the evaluation of
this strategy in more complex biological models of HIV infection.

CA 02818308 2013-05-16
13
As stated above, the invention is directed to "bifunctional" molecules, the
inventive
molecules being "bifunctional" in that they possess a pathogen binding
terminus (PBT) and
an antibody recruiting terminus (ABT) connected by a linker. The PBT is
designed to bind to
the HIV glycoprotein gp120 (gp120 on the viral membrane as well as gp120
displayed on
infected cells). The ABT is designed to bind antibodies and therefore redirect
endogenous
antibodies and hence the immune response to the pathogen. Formation of a
ternary complex
between these molecules, the antibodies, and the target pathogen, leads to
targeted
cytotoxicity through various mechanisms including antibody dependent cellular
cytotoxicity
(ADCC), or complement-dependent cytotoxicity (CDC).
The present invention is directed to pharmaceutical compositions comprising
the
above-described bifunctional molecules that can inhibit HIV entry into a
target cell, while
also recruiting antibodies to attack the HIV or an HIV infected cell, in a
pharmaceutically
acceptable carrier. As an aspect of the invention, therefore, we provide a
pharmaceutical
composition comprising a bifunctional molecule compound of the invention in
association
with a pharmaceutically acceptable carrier or excipient, adapted for use in
human or
veterinary medicine. Such compositions may be presented for use in
conventional manner in
admixture with one or more physiologically acceptable carriers or excipients.
The
compositions may optionally further contain one or more other therapeutic
agents which may,
if desired, be a different antiviral agent.
The bifunctional molecule compounds according to the invention may be
formulated
for oral, buccal, nasal, parenteral, topical or rectal administration, among
others, as otherwise
described herein.
In particular, the bifunctional compounds according to the invention may be
formulated for injection or for infusion and may be presented in unit dose
form in ampoules
or in multi-dose containers with an added preservative. The compositions may
take such
forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and
may contain
formulatory agents such as suspending, stabilizing and/ or dispersing agents.
Alternatively,
the active ingredient may be in powder form for constitution with a suitable
vehicle, e.g.
sterile, pyrogen-free water, before use.

CA 02818308 2013-05-16
14
The pharmaceutical compositions according to the invention may also contain
other
active ingredients such as antimicrobial agents, or preservatives.
The compositions may contain from 0.001-99% of the active material.
The invention further provides a process for preparing a pharmaceutical
composition
which comprises bringing a bifunctional molecule compound of the invention
into
association with a pharmaceutically acceptable excipient or carrier.
For administration by injection or infusion, dosages and desired drug
concentrations
of the disclosed pharmaceutical compositions may vary depending on the
particular use,
patient condition, age, drug tolerance, etc., as would be understood by one
skilled in the field.
Consequently, the determination of an appropriate dosage and/or route of
administration is
well within the skill of an ordinary practitioner, and the compounds can
certainly be
formulated without undue experimentation for administration in the treatment
of humans, for
example, using standard and well known dose-response protocols.
The amount of compound in a pharmaceutical composition of the instant
invention
that may be combined with the carrier materials to produce a single dosage
form will vary
depending upon the host and disease treated, the particular mode of
administration.
Preferably, the compositions should be formulated to contain between about
0.05 milligram
to about 750 milligrams or more, more preferably about 1 milligram to about
600 milligrams,
and even more preferably about 10 milligrams to about 500 milligrams of active
ingredient,
alone or in combination with at least one other ARM-HI compound which may be
used to
treat HIV infection or a secondary effect or condition thereof.
Brief Description Of The Drawings
Fig. 1 illustrates the effects of bifunctional ARM-HI compounds in forming a
ternary
complex between gp120 and an antibody.
Fig. 2 illustrates the dual mechanism of action exhibited by the bifunctional
molecules
of the present invention.
Fig. 3 shows Scheme 1, providing the synthesis of compound 2, presented in the
examples of the present application.

CA 02818308 2013-05-16
Fig. 4 shows Scheme 2, providing the synthesis of compound 9, presented in the
examples of the present application.
Fig. 5 shows Scheme 3, which provides for the general synthetic route to
analogous
compounds according to the present invention.
Fig. 6 shows the four compounds which were tested in a viral inhibition assay
and
compared to the inhibition in the same assay of a bifunctional compound (C5-
furan)
according to the present invention, also presented in Fig. 6.
Detailed description of the invention
The following terms are used to describe the present invention. In instances
where a
term is not specifically defined herein, that term is given an art-recognized
meaning by those
of ordinary skill applying that term in context to its use in describing the
present invention.
Where a range of values is provided, it is understood that each intervening
value, to
the tenth of the unit of the lower limit unless the context clearly dictates
otherwise (such as in
the case of a group containing a number of carbon atoms in which case each
carbon atom
number falling within the range is provided), between the upper and lower
limit of that range
and any other stated or intervening value in that stated range is encompassed
within the
invention. The upper and lower limits of these smaller ranges may
independently be included
in the smaller ranges is also encompassed within the invention, subject to any
specifically
excluded limit in the stated range. Where the stated range includes one or
both of the limits,
ranges excluding either both of those included limits are also included in the
invention.
The term "compound", as used herein, unless otherwise indicated, refers to any
specific chemical compound disclosed herein and includes tautomers,
regioisomers,
geometric isomers, and where applicable, optical isomers (enantiomers)
thereof, as well as
pharmaceutically acceptable salts and derivatives (including prodrug forms)
thereof. Within
its use in context, the term compound generally refers to a single compound,
but also may
include other compounds such as stereoisomers, regioisomers and/or optical
isomers
(including racemic mixtures) as well as specific enantiomers or
enantiomerically enriched
mixtures of disclosed compounds. The term also refers, in context to prodrug
forms of
compounds which have been modified to facilitate the administration and
delivery of
compounds to a site of activity. It is noted that in describing the present
compounds,

CA 02818308 2013-05-16
16
numerous substituents, linkers and connector molecules and variables
associated with same,
among others, are described. It is understood by those of ordinary skill that
molecules which
are described herein are stable compounds as generally described hereunder.
"Alkyl" refers to a fully saturated monovalent radical containing carbon and
hydrogen, and which may be cyclic, branched or a straight chain. Examples of
alkyl groups
are methyl, ethyl, n-butyl, sec-butyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, n-
decyl, isopropyl,
2-methylpropyl, cyclopropyl, cyclopropylmethyl, cyclobutyl, cyclopentyl,
cyclopentylethyl,
cyclohexylethyl and cyclohexyl, among others. Preferred alkyl groups are Ci-C6
or Ci-C3
alkyl groups.
"Aryl" or "aromatic", in context, refers to a substituted (as otherwise
described
herein) or unsubstituted monovalent aromatic radical having a single ring
(e.g., benzene or
phenyl) or condensed rings (e.g., naphthyl, anthracenyl, phenanthrenyl, etc.)
and can be
bound to the compound according to the present invention at any available
stable position on
the ring(s) or as otherwise indicated in the chemical structure presented.
Other examples of
aryl groups, in context, may include heterocyclic aromatic ring systems
"heteroaryl" groups
having one or more nitrogen, oxygen, or sulfur atoms in the ring (moncyclic)
such as
imidazole, furyl, pyrrole, furanyl, thiene, thiazole, pyridine, pyrimidine,
pyrazine, thazole,
oxazole or fused ring systems such as indole, quinoline, etc., among others,
which may be
optionally substituted as described above. Among the heteroaryl groups which
may be
mentioned include nitrogen-containing heteroaryl groups such as pyrrole,
pyridine, pyridone,
pyridazine, pyrimidine, pyrazine, pyrazole, imidazole, triazole, triazine,
tetrazole, indole,
isoindole, indolizine, purine, indazole, quinoline, isoquinoline, quinolizine,
phthalazine,
naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine,
imidazopyridine,
imidazotriazine, pyrazinopyridazine, acridine, phenanthridine, carbazole,
carbazoline,
perimidine, phenanthroline, phenacene, oxadiazole, benzimida7ole,
pyrrolopyridine,
pyrrolopyrimidine and pyridopyrimidine; sulfur-containing aromatic
heterocycles such as
thiophene and benzothiophene; oxygen-containing aromatic heterocycles such as
furan,
pyran, cyclopentapyran, benzofuran and isobenzofuran; and aromatic
heterocycles
comprising 2 or more hetero atoms selected from among nitrogen, sulfur and
oxygen, such as
thiazole, thiadizole, isothiazole, benzoxazole, benzothiazole,
benzothiadiazole,
phenothiazine, isoxazole, furazan, phenoxazine, pyrazoloxazole,
imidazothiazole,

CA 02818308 2013-05-16
17
thienofuran, furopyrrole, pyridoxazine, furopyridine, furopyrimidine,
thienopyrimidine and
oxazole, among others.
Alternative aryl and heteroaryl groups according to the present invention
preferably
include, for example, phenyl, naphthyl, pyridyl (2-, 3- or 4-pyridyl group),
thiazolyl (2-, 4- or
5-thiazole), isothiazolyl, oxazolyl (2-, 4- or 5-oxazole), isoxazolyl, furanyl
(2- or 3-furan) or
thiophenyl (2- or 3-thiophene). Monocyclic and bicyclic aryl and heteroayl
groups are as
otherwise described herein.
In alternative embodiments, preferred heteroaryl groups are 5- or 6-membered
aryl or
heteroaryl group according to the chemical structure:
v\-- - ---- - '''--- \- - I= :-.-. N - .\- - - .... -- ' -
- -.- = - . . v \ " " . ' - '. - ...- - = , . - -1 \ - -'''.
z, , XN , NN 'wA X X
N
W W , w N , w N ,
,
Y---------A, , \\--------) , N-------'--
-7-õzõ,/- ,
W W W
\
-Csrjr-
cX
W W'
' W
r-\ W
%__W
vv
,N1¨
-N i¨
N.:-,--õ_ _ / NI /
Where W is H, -(CH2)õOH, -(CH2)nCOOH, Ci-C6 alkyl,
-(CH2)õ0-(C1-C6 alkyl), -(CH2)nC(0)-(C1-C6 alkyl), -(CH2),NIC(0)-R1, -
(CH2)õC(0)-
NRIR2, ¨(CH20)110H, -(CH20)nCOOH, Ci-C6 alkyl, -(CH20)n0-(C1-C6 alkyl),

CA 02818308 2013-05-16
18
-(CH20)õC(0)-(C1-C6 alkyl), -(CH20)nNHC(0)-R1, -(CH20)õC(0)-NR1R2,NO2, CN,
halogen
(F, Cl, Br, I, preferably F or Cl) or a monocyclic aryl or heteroaryl group
which itself is
optionally substituted (especially an optionally substituted benzoyl or benzyl
group);
W' is I-1, -(CH2)OH, -(CH2)nCOOH, C1-C6 alkyl, -(CH2)0-(C1-C6 alkyl) or
halogen
(preferably F or Cl); and
Y is 0, S or N-R, where R is H or a C1-C3 alkyl group.
In still other embodiments, preferred aryl or heteroaryl groups include those
which are
substituted according to the chemical structures:
µ/1/111' vdtil
1µ1
, NLZ.
N
w2 w2 N N w2 N w2 N
w2
fµisr= csssf=-
\/\
c -1¨W2
/W2 Xw 2
wN12
, X
X
,f1Nr*. X w2
X
NIzsis\ rN\N¨X
N,- N
N
Where W2 is H, -(CH2)0H, -(CH2)nCOOH, C1-C6 alkyl, -(CH2)õ0-(C1-C6 alkyl),
-(CH2)nC(0)-(C1-C6 alkyl), -(CH2)nNHC(0)-R1, -(CH2),C(0)-NR1R2, -(CH20)n0H,
-(CH20)nC0011, C1-C6 alkyl, -(CH20)n0-(C1-C6 alkyl), -(CFI20)nC(0)-(C1-C6
alkyl),
-(CH20)nNHC(0)-R1, -(CH20)nC(0)-N121122,NO2, CN or halogen (preferably F or
Cl);
X is a group ¨NH-, -NHC(0)-, -0-, -S-, -S(0)-, SO2- or ¨NH-C(0)-NH-; and
Y is 0, S or N-R, where R is H or a C1-C3 alkyl group.
The term "substituted" shall mean substituted at a carbon (or nitrogen)
position within
context, hydroxyl, carboxyl, cyano (CN), nitro (NO2), halogen (preferably, 1,
2 or 3

CA 02818308 2013-05-16
19
halogens, especially on an alkyl, especially a methyl group such as a
trifluoromethyl), alkyl
group (preferably, C1-C10, more preferably, Ci-C6), aryl (especially phenyl
and substituted
phenyl for example benzyl or benzoyl), alkoxy group (preferably, C1-C6 alkyl
or aryl,
including phenyl and substituted phenyl), ester (preferably, C1-C6 alkyl or
aryl) including
alkylene ester (such that attachment is on the alkylene group, rather than at
the ester function
which is preferably substituted with a Ci-C6 alkyl or aryl group), preferably,
C1-C6 alkyl or
aryl, halogen (preferably, F or Cl), nitro or amine (including a five- or six-
membered cyclic
alkylene amine, further including a C1-C6 alkyl amine or Ci-C6 dialkyl amine
which alkyl
groups may be substituted with one or two hydroxyl groups), amido, which is
preferably
substituted with one or two C1-C6 alkyl groups (including a carboxamide which
is substituted
with one or two Ci-C6 alkyl groups), alkanol (preferably, C1-C6 alkyl or
aryl), or alkanoic
acid (preferably, C1-C6 alkyl or aryl). The term "substituted" shall also mean
within its
context of use alkyl, alkoxy, halogen, amido, carboxamido, keto, carboxy,
ester, keto, nitro,
cyano and amine (especially including mono- or di- C1-C6 alkyl substituted
amines which
may be optionally substituted with one or two hydroxyl groups). In certain
embodiments
preferred substituents will include for example, ¨NH-, -NHC(0)-, -0-, -(CH2)m-
(m and n are
at least 1 as otherwise described herein), -S-, -S(0)-, SO2- or ¨NH-C(0)-NH-, -
(CH2)0H,
-(CH2)COOH, C1-C6 alkyl, -(CH2)n0-(C1-C6 -(CH2)nC(0)-(C1-C6
-(CH2)õNHC(0)-R1, -(CH2)õC(0)-NRIR2, -(CH20).0H, -(CH20)nCOOH, C1-C6 alkyl,
-(OCH2)n0-(C1-C6 alkyl), -(OCH2)nC(0)-(C1-C6 alkyl), -(OCH2)nNHC(0)-R1,
-(CH20)C(0)-NRIR2,NO2, CN or halogen (F, Cl, Br, I, preferably F or Cl),
depending on
the context of the use of the substituent.
Any substitutable position in a compound according to the present invention
may be
substituted in the present invention, but no more than 3, more preferably no
more than 2
substituents (in some instances only 1 or no substituents) is present on a
ring. Preferably, the
term "unsubstituted" shall mean substituted with one or more H atoms.
The term "patient" or "subject" is used throughout the specification within
context to
describe an animal, generally a mammal and preferably a human, to whom
treatment,
including prophylactic treatment (prophylaxis), with the compositions
according to the
present invention is provided. For treatment of those infections, conditions
or disease states
which are specific for a specific animal such as a human patient or a patient
of a particular
gender, such as a human male patient, the term patient refers to that specific
animal.
Compounds according to the present invention are useful for treating and/or
reducing the

CA 02818308 2013-05-16
likelihood of HIV infections or the secondary effects of HIV infections,
especially including
AIDS and/or ARC.
The term "effective" is used herein, unless otherwise indicated, to describe
an
amount of a compound or composition which, in context, is used to produce or
effect an
intended result, whether that result relates to the inhibition of the effects
of a toxicant on a
subject or the treatment of a subject for secondary conditions, disease states
or
manifestations of exposure to toxicants as otherwise described herein. This
term subsumes
all other effective amount or effective concentration terms (including the
term
"therapeutically effective") which are otherwise described in the present
application.
The terms "treat", "treating", and "treatment", etc., as used herein, refer to
any action
providing a benefit to a patient at risk for HIV infection or having an HIV
infection,
including improvement in the condition through lessening or suppression of
titers of HIV or
at least one symptom of HIV, prevention or delay in progression of the
disease, prevention or
delay in the onset of disease states or conditions which occur secondary to
HIV, including
AIDS or ARC, among others. Treatment, as used herein, encompasses both
prophylactic
and therapeutic treatment. The term "prophylactic" when used, means to reduce
the
likelihood of an occurrence or the severity of an occurrence within the
context of the
treatment of HIV, as otherwise described hereinabove.
The term "human immunodeficieincy virus" or "HIV" shall be used to describe
human immunodeficiency viruses 1 and 2 (HIV-1 and HIV-2), the growth or
replication of
which may be inhibited or disease states of which may be treated using one or
more methods
according to the present invention. Viruses which may be treated according to
the present
invention include, for example, human immunodeficiency viruses 1 and 2 (HIV-1
and HIV-
2), among others. The term HIV includes mutant strains of HIV including "drug
resistant" or
"multiple drug resistant" strains of the HIV virus which have mutated to be
resistant to one or
more clinically approved anti-HIV agents, including, in particular, HIV
strains which are
resistant to one or more NRTI compounds and/or NNRTI compounds. Exemplary HIV
drug
resistant strains which may be effectively treated using compounds according
to the present
invention include the following, among others: (defined by their reverse
transcriptase or RT
mutation)- XXBRU, K65R, Y115F, F116Y, Q151M, M184V, L74V, V75T, 4XZT, T215Y,
K103N, T215Y/M184V, 5705-72, 488-101, C910-6, LA1M184V, G910-6 L100I, K101E,

CA 02818308 2013-05-16
21
K103N, V106A, D1 10E, V179D, Y181C, D185E, D186E, Y188H, G190E, E138K, M41L,
D67N, K7OR, T215Y/F, K219Q/E, Y181C, K103N, L100I, Y188C/H, among others,
including HIV-1 isolates JR-FL, ADA, HXBc2, SF162 and BaL, among others.
The terms "ARC" and "AIDS" refer to syndromes of the immune system caused by
the human immunodeficiency virus, which are characterized by susceptibility to
certain
diseases and T cell counts which are depressed compared to normal counts. HIV
progresses
from Category 1 (Asymptomatic HIV Disease) to Category 2 (ARC), to Category 3
(AIDS),
with the severity of the disease.
A Category 1 HIV infection is characterized by the patient or subject being
HIV
positive, asymptomatic (no symptoms) and having never had fewer than 500 CD4
cells. If
the patient has had any of the AIDS-defining diseases listed for categories 2
(ARC) or 3
(AIDS), then the patient is not in this category. If the patient's t-cell
count has ever dropped
below 500, that patient is considered either Category 2 (ARC) or Category 3
(AIDS).
A Category 2 (ARC) infection is characterized by the following criteria: The
patient's T-cells have dropped below 500 but never below 200, and that patient
has never had
any Category 3 diseases (as set forth below) but have had at least one of the
following
defining illnesses --
Bacillary angiomatosis
Candidiasis, oropharyngeal (thrush)
Candidiasis, vulvovaginal; persistent, frequent, or poorly responsive to
therapy
Cervical dysplasia (moderate or severe)/cervical carcinoma in situ
Constitutional symptoms, such as fever (38.5 C) or diarrhea lasting longer
than 1
month
Hairy leukoplakia, oral
Herpes zoster (shingles), involving at least two distinct episodes or more
than one
dermatome
Idiopathic thrombocytopenic purpura
Listeriosis
Pelvic inflammatory disease, particularly if complicated by tubo-ovarian
abscess
Peripheral neuropathy

CA 02818308 2013-05-16
22
According to the U.S. government, in Category 2 ARC, the immune system shows
some signs of damage but it isn't life-threatening.
A Category 3 (AIDS) infection is characterized by the following criteria:
T-cells have dropped below 200 or the patient has had at least one of the
following defining
illnesses --
Brain Toxoplasmosis
Candidiasis of bronchi, trachea, or lungs
Candidiasis, esophageal
Cervical cancer, invasive**
Coccidioidomycosis, disseminated or extrapulmonary
Cryptococcosis, extrapulmonary
Cryptosporidiosis, chronic intestinal (greater than 1 month's duration)
Cytomegalovirus disease (other than liver, spleen, or nodes)
Cytomegalovirus retinitis (with loss of vision)
Encephalopathy, HIV-related
Herpes simplex: chronic ulcer(s) (greater than 1 month's duration); or
bronchitis,
pneumonitis, or esophagitis
Histoplasmosis, disseminated or extrapulmonary
Isosporiasis, chronic intestinal (greater than 1 month's duration)
Kaposi's sarcoma
Lymphoma, Burkitt's (or equivalent term)
Lymphoma, immunoblastic (or equivalent term)
Lymphoma, primary, of brain
Mycobacterium avium complex or M. kansasii, disseminated or extrapulmonary
Mycobacterium tuberculosis, any site (pulmonary** or extrapulmonary)
Mycobacterium, other species or unidentified species, disseminated or
extrapulmonary
Pneumocystis carinii pneumonia
Pneumonia, recurrent
Progressive multifocal leukoencephalopathy
Salmonella septicemia, recurrent
Wasting syndrome due to HIV

CA 02818308 2013-05-16
23
The term "coadministration" or "combination therapy" shall mean that at least
two
compounds or compositions are administered to the patient at the same time,
such that
effective amounts or concentrations of each of the two or more compounds may
be found in
the patient at a given point in time. Although compounds according to the
present invention
may be co-administered to a patient at the same time, the term embraces both
administration
of two or more agents at the same time or at different times, provided that
effective
concentrations of all coadministered compounds or compositions are found in
the subject at a
given time. In certain preferred aspects of the present invention, one or more
of the
bifunction ARM-HI compounds described above, are coadministered in combination
with at
least one additional anti-HIV agent as otherwise described herein in a
cocktail for the
treatment of HIV infections. In particularly preferred aspects of the
invention, the co-
administration of compounds results in synergistic anti-HIV activity of the
therapy.
The term "additional anti-HIV agent" shall mean a traditional anti-HIV agent
(ie., a
non-bifunctional ARM-HI compound as otherwise described herein) which may be
co-
administered to a patient along with ARM-HI compounds according to the present
invention
in treating a patient for HIV. Such compounds include, for example, agents
such as
nucleoside reverse transcriptase inhibitors (NRTI), non-nucloeoside reverse
transcriptase
inhibitors, protease inhibitors and fusion inhibitors. Exemplary compounds
include, for
example, Amprenivir, Abacavir, Acemannan, Acyclovir, AD-439, AD-519, Adefovir
dipivoxil, Alpha Interferon, Ansamycin, 097, AR 177, Beta-fluoro-ddA, BMS-
232623 (CGP-
73547), BMS-234475 (CGP-61755), CI-1012, Cidofovir, Curdlan sulfate,
Cytomegalovirus
Immune globin, Ganciclovir, Dideoxyinosine, DMP-450, Efavirenz (DMP-266),
EL10,
Famciclovir, FTC, GS 840, HBY097, Hypericin, Recombinant Human Interferon
Beta,
Interferon alfa-n3, Indinavir, ISIS-2922, KNI-272, Lamivudine (3TC),
Lobucavir, Nelfinavir,
Nevirapine, Novapren, Peptide T Octapeptide Sequence, Trisodium
Phosphonoformate,
PNU-140690, Probucol, RBC-CD4, Ritonavir, Saquinavir, Valaciclovir, Virazole
Ribavirin,
VX-478, Zalcitabine, Zidovudine (AZT), Tenofovir diisoproxil fumarate salt,
Combivir,
Abacavir succinate, T-20), AS-101, Bropirimine, CL246, ELIO, FP-21399, Gamma
Interferon, Granulocyte Macrophage Colony Stimulating Factor (GM-CSF), HIV
Core
Particle Immunostimulant, Interleukin-2 (IL-2), Immune Globulin Intravenous,
IMREG-1,
IMREG-2, Imuthiol Diethyl Dithio Carbamate, Alpha-2 Interferon, Methionine-
Enkephalin,
MTP-PE (Muramyl-Tripeptide), Granulocyte Colony Stimulating Factor (GCSF),
Remune,
rCD4 (Recombinant Soluble Human CD4-IgG), rCD4-IgG Hybrids, Recombinant
Soluble

CA 02818308 2013-05-16
24
Human CD4, Interferon Alfa 2a, SK&F1-6528, Soluble T4, Thymopentin, Tumor
Necrosis
Factor (TNF), AK602, Alovudine, Amdoxovir, AMD070, Atazanavir (Reyataz),
AVX754
(apricitabine), Bevirimat, B1-201, BMS-378806, BMS-488043, BMS-707035, C31G,
Carbopol 974P, Calanolide A, Carrageenan, Cellulose sulfate, Cyanovirin-N,
Darunavir,
Delavirdine, Didanosine (Videx), Efavirenz, Elvucitabine, Emtricitabine,
Fosamprenavir
(Lexiva), Fozivudine tidoxil, GS 9137, GSK-873,140 (aplaviroc), GSK- 364735,
GW640385
(brecanavir), HG0004, HGTV43, INCB9471, KP-1461, Lopinavir, Mifepristone
(VGX410),
MK-0518, PPL-100, PRO 140, PRO 542, PRO 2000, Racivir, SCH-D (vicriviroc),
SPO1A,
SPL7013, TAK-652, Tipranavir (Aptivus), TNX-355, TMC125 (etravirine), UC-781,
UK-
427,857
(Maraviroc)http://aidsinfo.nih.gov/DrugsNew/DrugDetailNT.aspx?MenuItem4)rugs&Se
arc
h 4>n&int id =408, Valproic acid, VRX496, Zalcitabine, Valganciclovir,
Clindamycin with
Primaquine, Fluconazole Pastille, Nystatin Pastille, Eflomithine, Pentamidine,
Isethionate,
Trimethoprim, Trimethoprim/sulfa, Piritrexim, Pentamidine isethionate,
Spiramycin,
Intraconazole-R51211, Trimetrexate, Daunorubicin, Recombinant Human
Erythropoietin,
Recombinant Human Growth Hormone, Megestrol Acetate, Testosterone, Aldesleukin
(Proleukin), Amphotericin B, Azithromycin (Zithromax), Calcium hydroxyapatite,
Doxorubicin, Dronabinol, Entecavir, Epoetin alfa, Etoposide, Fluconazole,
Isoniazid,
Itraconazole (Sporanox), Megestrol, Paclitaxel (Taxol), Peginterferon alfa-2,
Poly-L-lactic
acid (Sculptra), Rifabutin (Mycobutin), Rifampin, Somatropin and
Sulfamethoxazole/Trimethoprim. Preferred anti-HIV compounds for use in the
present
invention include, for example, 3TC (Lamivudine), AZT (Zidovudine), (-)-FTC,
ddI
(Didanosine), ddC (zalcitabine), abacavir (ABC), tenofovir (PMPA), D-D4FC
(Reverset),
D4T (Stavudine), Racivir, L-FddC, L-FD4C, NVP (Nevirapine), DLV (Delavirdine),
EFV
(Efavirenz), SQVM (Saquinavir mesylate), RTV (Ritonavir), IDV (Indinavir), SQV
(Saquinavir), NFV (Nelfmavir), APV (Amprenavir), LPV (Lopinavir), fusion
inhibitors such
as T20, among others, fuseon and mixtures thereof
The term "pharmaceutically acceptable salt" is used throughout the
specification to
describe a salt form of one or more of the compounds herein which are
presented to increase
the solubility of the compound in saline for parenteral delivery or in the
gastric juices of the
patient's gastrointestinal tract in order to promote dissolution and the
bioavailability of the
compounds. Pharmaceutically acceptable salts include those derived from
pharmaceutically
acceptable inorganic or organic bases and acids. Suitable salts include those
derived from

CA 02818308 2013-05-16
alkali metals such as potassium and sodium, alkaline earth metals such as
calcium,
magnesium and ammonium salts, among numerous other acids well known in the
pharmaceutical art. Sodium and potassium salts may be particularly preferred
as
neutralization salts of carboxylic acid containing compositions according to
the present
invention. The term "salt" shall mean any salt consistent with the use of the
compounds
according to the present invention. In the case where the compounds are used
in
pharmaceutical indications, including the treatment of HIV infections, the
term "salt" shall
mean a pharmaceutically acceptable salt, consistent with the use of the
compounds as
pharmaceutical agents.
The term "antibody binding terminal moiety", "antibody binding terminus" or
"antibody binding moiety" (ABT within the general formula of compounds
according to the
present invention) is used to describe that portion of a bifunctional ARM-HI
compound
according to the present invention which comprises at least one small molecule
or hapten
which can bind to antibodies within the patient. The term "hapten" is used to
describe a
small-molecular-weight inorganic or organic molecule that alone is not
antigenic but which
when linked to another molecule, such as a carrier protein (albumin, etc.) or
in the case of the
present invention, as an antibody terminus in the present compounds, is
antigenic; and an
antibody raised against the hapten (generally, the hapten bonded or complexed
to the carrier)
will react with the hapten alone. Because, in many instances, anti-hapten
(anti-DNP)
antibodies are already present in the human blood stream as endogenous
antibodies because
they naturally become raised to endogenous haptens (already present in
patients), no pre-
vaccination is necessary for ARM-HI activity.
It is preferred that the antibody binding terminal comprise a hapten which is
reactive
with (binds to) an endogenous antibody that pre-exists in the patient prior to
initiate therapy
with the compounds of the present invention and does not have to be separately
raised as part
of a treatment regimen (for example, by vaccination or other approach for
enhancing
immunogenicity). Thus, haptens which comprise a di-or trinitro phenyl group as
depicted
below, or a digalactose hapten (Gal-Gal-Z, preferably Gal-Gal-sugar,
preferably Gal-Gal-
Glu), are preferred. Additionally, a compound according to the general
structure:

CA 02818308 2013-05-16
26
O0 0
X" ________________________________________________
Where X" is 0, CH2, Me, S; and
RI is H, a C1-C3 alkyl group or a -C(0)(C1-C3) group;
May be used as haptens in the present invention.
Further, a moiety according to the chemical structure:
p-N
N NO2
Xb
Where Xb is a bond, 0, CH2, NR' or S may also be used as a hapten (ABT) in the
present
invention.
Other ABT moieties include the following structures:
0 0
02N
s. NO2
IP NO2 H
HR, HN
H
NO2 NO2
HN
Hu' NO2
HWNO,
H "Nyw,mõ NO2
NO2 i4
0 0
Each of the above amino acid ABT moieties may be further substituted with a
dinitrophenyl
group through an X group, e.g., CH2- , sulfoxide, sulfone, etc. group as
otherwise described
herein to provide the following ABT moieties:

CA 02818308 2013-05-16
27
DNP
I NH ;SSS'NHN
DNP
Hrv
HIT1
DNP
DNP
DNP\NH
DNP
HN
DNP NH
y/\/1,(DNP
0
In the above structures in each of the molecules (with the exception of the
first, which
is DNP amine), DNP may be linked to the structure where the NO2 is linked.
The di- or trinitro phenyl hapten (ABT) moiety for use in the present
invention
(Dinitropheny or DNP hapten is preferred) may be represented by the following
formula:
NO2
NO2
401
Y'
Where Y' is H or NO2 (preferably H);
X is 0, CH2, NR', S(0), S(0)2, -S(0)20, -OS(0)2, or OS(0)20; and
R' is H, a C1-C3 alkyl group, or a -C(0)(C1-C3) group.
The (Gal-Gal-Z) hapten is represented by the chemical formula:
HO OH
H_o H H 0
0 H
H 0 .
0 H H_o
0 H
Where X' is CH2, 0, or S, preferably 0;

CA 02818308 2013-05-16
28
Ry is H or C1-C3 alkyl; and
Z is a bond, a monosaccharide, disaccharide, oligosaccharide, glycoprotein or
glycolipid,
preferably a sugar group, more preferably a sugar group selected from the
monosaccharides,
including aldoses and ketoses, and disaccharides, including those
disaccharides described
herein. Monosaccharide aldoses include monosaccharides such as aldotriose (D-
glyceraldehdye, among others), aldotetroses (D-erythrose and D-Threose, among
others),
aldopentoses, (D-ribose, D-arabinose, D-xylose, D-Iyxose, among others),
aldohexoses (D-
allose, D-altrose, D-Glucose, D-Marmose, D-gulose, D-idose, D-galactose and D-
Talose,
among others), and the monosaccharide ketoses include monosaccharides such as
ketotriose
(dihydroxyacetone, among others), ketotetrose (D-erythmlose, among others),
ketopentose
(D-ribulose and D-xylulose, among others), ketohexoses (D-Psicone, D-Fructose,
D-Sorbose,
D-Tagatose, among others), aminosugars, including galactoseamine , sialic
acid, N-
acetylglucosamine, among others and sulfosugars, including sulfoquinovose,
among others.
Exemplary disaccharides which find use in the present invention include
sucrose (which may
have the glucose optionally N-acetylated), lactose (which may have the
galactose and/or the
glucose optionally N-acetylated), maltose (which may have one or both of the
glucose
residues optionally N-acetylated), trehalose (which may have one or both of
the glucose
residues optionally N-acetylated), cellobiose (which may have one or both of
the glucose
residues optionally N-acetylated), kojibiose (which may have one or both of
the glucose
residues optionally N-acetylated), nigerose (which may have one or both of the
glucose
residues optionally N-acetylated), isomaltose (which may have one or both of
the glucose
residues optionally N-acetylated), 0,13-trehalose (which may have one or both
of the glucose
residues optionally N-acetylated), sophorose (which may have one or both of
the glucose
residues optionally N-acetylated), laminaribiose (which may have one or both
of the glucose
residues optionally N-acetylated), gentiobiose (which may have one or both of
the glucose
residues optionally N-acetylated), turanose (which may have the glucose
residue optionally
N-acetylated), maltulose (which may have the glucose residue optionally N-
acetylated),
palatinose (which may have the glucose residue optionally N-acetylated),
gentiobiluose
(which may have the glucose residue optionally N-acetylated), mannobiose,
melibiose (which
may have the glucose residue and/or the galactose residue optionally N-
acetylated),
melibiulose (which may have the galactose residue optionally N-acetylated),
rutinose, (which
may have the glucose residue optionally N-acetylated), rutinulose and
xylobiose, among
others. Oligosaccharides for use in the present invention as Z can include any
sugar of three
or more (up to about 100) individual sugar (saccharide) units as described
above (i.e., any one

CA 02818308 2013-05-16
29
or more saccharide units described above, in any order, especially including
glucose and/or
galactose units as set forth above), or for example, fructo-oligosaccharides,
galactooligosaccharides and mannan-oligosaccharides ranging from three to
about ten-fifteen
sugar units in size. Glycoproteins for use in the present invention include,
for example, N-
glycosylated and 0-glycosylated glycoproteins, including the mucins,
collagens, transferring,
ceruloplasmin, major histocompatability complex proteins (MHC), enzymes,
lectins and
selectins, calnexin, calreticulin, and integrin glycoprotein Hb/lIa, among
others. Glycolipids
for use in the present invention include, for example, glyceroglycolipids
(galactolipids,
sulfolipids), glycosphingolipids, such as cerebrosides, galactocerebrosides,
glucocerebrosides
(including glucobicaranateoets), gangliosides, globosides, sulfatides,
glycophosphphingolipids and glycocalyx, among others.
Preferably, Z is a bond (linking a Gal-Gal disaccharide to a linker or
connector
molecule) or a glucose or glucosamine (especially N-acetylglucosamine).
It is noted that Z is linked to a galactose residue through a hydroxyl group
or an amine group
on the galactose of Gal-Gal, preferably a hydroxyl group. A preferred hapten
is Gal-Gal-Glu
which is represented by the structure:
HO OH
H FLO H HO
OH
HO H OH
0 FLO
0 H__co
0 = 0
HO ____________________________________________
xs
Where Xs is OH or NflAc.
Other ABT groups include, for example, the following groups:

CA 02818308 2013-05-16
0
XR-1-
0
HO +C)-1\0NMe3
HO
OH 0 Phosphoryl
Rhamnose Choline
0
0
Sel
0
0
Menadione
Carboxyethyl Lysine (CEL)
Where XR is 0 or S; and
Xm is 0 or S.
It is noted in the carboxyethyl lysine ABT moiety either one, two or three of
the
nitrogen groups may be linked to the remaining portion of the molecule through
the linker or
one or both of the remaining nitrogen groups may be substituted with a
dinitrophenyl through
an X group as otherwise described herein.
The term "pathogen binding terminus" or "pathogen binding terminal moiety"
("PBT") is use to described that portion of a difunctional ARM-HI compound
according to
the present invention which comprises at least one small molecule or moiety
which can bind
specifically to is capable of binding to gp120 envelope protein on HIV virus
or a cell surface
of CD4 cells which are infected with HIV (HIV -I) in said patient.

CA 02818308 2013-05-16
31
PBT groups (i.e., the chemical moiety connected to linkers and ABT in the
bifunctional chemical compound below) for use in the present invention include
those which
are found in the following bifunctional compounds having the following
chemical structure:
0 ARYL
x3)k
X2 0
Y3
\ 0
RN
RY
AB
ABT
Where _____ is an antibody binding terminus (moiety) comprising a hapten
which is
capable of binding to an antibody present in a patient (preferably a DNP
group);
LINKER
______ is a linker molecule which chemical links ABT to RY or directly to the
indole
moiety at the carbon atom to which RY is attached and which optionally
includes a connector
CT which may be a bond or a connector molecule;
ARYL
_____ is an aromatic or heteroaromatic group, preferably a monocyclic or
bicyclic
aromatic or heteroaromatic group;
RY is absent or is an optionally substituted aryl or heteroaryl group or 0,
(CH2)j, NR', -S-,
-NHC(0)-, -NHC(0)NH-, S(0), S(0)2, -S(0)20, -OS(0)2, or OS(0)20;
X2 is H, -(CH2)n0H, -(CH2)11COOH, C1-C6 alkyl, -(CH2).0-(C1-C6 alkyl),
-(CH2)nC(0)-(C1-C6 alkyl), -(CH2)nNHC(0)-R1, -(CH2)nC(0)-NRIR2, -(CH20)n0H,
-(CH20)COOH, C1-C6 alkyl, -(CH20)n0-(C1-C6 alkyl),
-(CH20).C(0)-(C1-C6 alkyl), -(CH20)nNHC(0)-R1, -(CH20)11C(0)-NRIR2,NO2, CN or
halogen (F, Cl, Br, I, preferably F or Cl);

CA 02818308 2013-05-16
32
X3 is H, -(CH2)60H, -(CH2)COOH, Ci-C6 alkyl, -(CH2),10-(C1-C6 alkyl),
-(CH2)6C(0)-(CI-C6 alkyl), -(CH2)nNHC(0)-R1, -(CH2)nC(0)-NR1R2, -(CH20).0H,
-(CH20)COOH, Ci-C6 alkyl, -(CH20)60-(C1-C6 alkyl), -(CH20)nC(0)-(C1-C6 alkyl),
-(CH20)õNHC(0)-R1, -(CH20)õC(0)-NRIR2,NO2, CN, halogen (F, Cl, Br, I,
preferably F or
Cl) or a monocyclic aryl or heteroaryl group which itself is optionally
substituted;
RI is H or a C1-C3 alkyl group;
R1 and R2 are each independently H or a C1-C6 alkyl group;
i is 0 or I, preferably 1;
j is 1,2 or 3;
k is 0, 1, 2 or 3, preferably 0, 1 or 2;
n is 0, 1, 2, 3, 4, 5, 6, preferably 0-3;
Y3 is H or a Ci-C3 alkyl group (preferably, disposed out of or into the plane,
preferably out of
the plane on the chiral carbon; and
RN is H or a Ci-C3 alkyl group which is optionally substituted with one or two
hydroxyl
groups or up to three halogen groups (preferably F)
or a pharmaceutically acceptable salt, enantiomer, solvate or polymorph
thereof.
Preferred PBT groups for use in the present invention include those (i.e., the
chemical
moiety connected to the linker and ABT below- connected to X) according to the
chemical
formula:

CA 02818308 2013-05-16
33
0 ARYL1
X2 0
i y3
\ 0
RN
ARYL2
X _______________ LINKER ______ ABT
ARYL1
Where ____________ is a monocyclic or bicyclic aryl or heteroaryl group
according to
the chemical structure:

CA 02818308 2013-05-16
34
w,
v\ N'\ N.'\ 'Q NN
V\ /: NV
( ( f (
' XN , NN AN , XN
W W 'W W
/
Lv I C I I
-..--,N,,, , ,.,,y,,---.Z.,.z....õ/ ,
W ' W V/
VW
W W W
xrPr.-\
NN
w w' , w w r w' w
,
w
,r=ss'sr"\ W
I----\,õ )=-----\--,N1 - I ------ NW W)--------\ -NI-
N----.... /
N N , N
Where W is H, -(CH2)õOH, -(CH2)nCOOH, Ci-C6 alkyl,
-(CH2)n0-(C1-C6 alkyl), -(CH2)õC(0)-(C1-C6 alkyl), -(CH2)nNHC(0)-R1, -
(CH2)nC(0)-
NRIR2, -(CH20)n0H, -(C1120)õCOOH, C1-C6 alkyl, -(CH20)n0-(C1-C6 alkyl), -
(CH20)nC(0)-(C1-C6 alkyl), -(CH20).NHC(0)-R1, -(CH20)nC(0)-NRIR2,NO2, CN,
halogen
(F, Cl, Br, I, preferably F or Cl) or a monocyclic aryl or heteroaryl group
which itself is
optionally substituted (especially an optionally substituted benzoyl or benzyl
group);
W' is H, -(CH2)n0H, -(CH2)nCOOH, Ci-C6 alkyl, -(CH2)n0-(C1-C6 alkyl) or
halogen
(preferably F or Cl);
ARYL2 1---X--
c' is a group according to chemical structure:

CA 02818308 2013-05-16
/
%AN
X1 N N N N
N = /
N 9 w2 N w2 N 9 w2 w2 N
w/2
_p\fspr= >f, >r= sr\s
(N
/\NI=w2 -1Y¨W2
X , X X
W2
X
r\-\x
N¨X \N¨X
N N /
Where W2 is H, -(CH2)n0H, -(CH2).COOH, C1-C6 alkyl, -(CH2)n0-(C1-C6 alkyl),
-(CH2).C(0)-(C1-C6 alkyl), -(CH2)nNHC(0)-R1, -(CH2)nC(0)-NRIR2, -(CH20)n0H,
-(CH20)nCOOH, C1-C6 alkyl, -(CH20).0-(C1-C6 alkyl), -(CH20)nC(0)-(C1-C6
alkyl),
-(CH20)õNHC(0)-R1, -(CH20)õC(0)-NRIR2,NO2, CN or halogen (preferably F or Cl);
X is a group ¨(CH2)nNH-, -(CH2)nNHC(0)-, -(CH2)n0-, -(CH2)m-, -(CH2)nS-, -
(CH2)nS(0)-,
ARYL2
-(CH2)n SO2- or ¨(CH2)nNH-C(0)-NH- which links __ to the linker;
Y is 0, S or N-R where R is H or a CI-C3 alkyl group;
X2 is H, -(CH2)n0H, -(CH2)nCOOH, Ci-C6 alkyl, -(CH2)n0-(C1-C6 alkyl),
-(CH2)nC(0)-(C1-C6 alkyl), -(CH2)nNHC(0)-R1, -(CH2)nC(0)-NR1R2, -(CH20)n0H,
-(CH20)nCOOH, Ci-C6 alkyl, -(CH20)n0-(C1-C6 alkyl),
-(CH20)nC(0)-(C1-C6 alkyl), -(CH20)nNHC(0)-R1 or -(CH20)11C(0)-NRIR2,NO2;
R1 and R2 are each independently H or a Ci-C6 alkyl group;
Y3 is H or a Ci-C3 alkyl group (preferably, disposed out of or into the plane,
preferably out of
the plane on the chiral carbon; and
RN is H or a C1-C3 alkyl group which is optionally substituted with one or two
hydroxyl
groups or up to three halogen groups (preferably F);
i is 0 or 1, preferably 1; and
m is 1, 2, 3, 4 or 5, preferably 1, 2 or 3, more preferably 1; and

CA 02818308 2013-05-16
36
Each n is independently 0, 1,2 or 3, or
a pharmaceutically acceptable salt, enantiomer, solvate or polymorph thereof.
The term "linker" refers to a chemical entity connecting an antibody binding
terminus
(ABT) moiety to a pathogen binding terminus (CBT) moiety, optionally through a
connector
moiety (CT) through covalent bonds. The linker between the two active portions
of the
molecule, that is the antibody binding terminus (ABT) and the pathogen binding
terminus
(PBT) ranges from about 5A to about 50A or more in length, about 6A to about
45A in
length, about 7A to about 40A in length, about 8A to about 35A in length,
about 9A to about
30A in length, about 10A to about 25A in length, about 7A to about 20 A in
length, about 5A
to about 16A in length, about 5A to about 15A in length, about 6A to about 14A
in length,
about 10A to about 20A in length, about 11A to about 25A in length, etc.
Linkers which are
based upon ethylene glycol units and are between 4 and 14 glycol units in
length may be
preferred. By having a linker with a length as otherwise disclosed herein, the
ABT moiety
and the PBT moiety may be situated to advantageously take advantage of the
biological
activity of compounds according to the present invention which bind to HIV
envelope protein
gp120 (gp120) and attract endogenous antibodies to the virus and/or infected
cells (e.g. HIV
infected CD4 cels) to which the compounds are bound, resulting in the
selective and targeted
death of those viruses and/or cells. The selection of a linker component is
based on its
documented properties of biocompatibility, solubility in aqueous and organic
media, and low
immunogenicity/antigenicity. Although numerous linkers may be used as
otherwise
described herein, a linker based upon polyethyleneglycol (PEG) linkages,
polypropylene
glycol linkages, or polyethyleneglycol-co-polypropylene oligomers (up to about
100 units,
about 1 to 100, about 1 to 75, about 1 to 60, about 1 to 50, about 1 to 35,
about 1 to 25, about
1 to 20, about 1 to 15,2 to 10, about 4 to 12, about 1 to 8, 1 to 3, 1 to 4,2
to 6, 1 to 5, etc.)
may be favored as a linker because of the chemical and biological
characteristics of these
molecules. The use of polyethylene (PEG) linkages is preferred. Alternative
preferred
linkers may include, for example, polyproline linkers and/or collagen linkers
as depicted
below (n is about 1 to 100, about 1 to 75, about 1 to 60, about 1 to 50, about
1 to 45, about 1
to 35, about 1 to 25, about 1 to 20, about 1 to 15, 2 to 10, about 4 to 12,
about 5 to 10, about 4
to 6, about 1 to 8, about 1 to 6 ,about 1 to 5, about 1 to 4, about 1 to 3,
etc.).

CA 02818308 2013-05-16
37
0
_ n polyproline linker
HO
0
11=1\
0 0
Or ¨ n collagen linker.
Preferred linkers include those according to the chemical structures:
R a
R3
03 A:10 N..s.S-C.Nr
N
or 0 m
Or a polypropylene glycol or polypropylene-co-polyethylene glycol linker
having between 1
and 100 glycol units;
Where Ra is H, C1-C3 alkyl or alkanol or forms a cyclic ring with R3 (proline)
and R3 is a side
chain derived from an amino acid preferably selected from the group consisting
of alanine
(methyl), arginine (propyleneguanidine), asparagine (methylenecarboxyamide),
aspartic acid
(ethanoic acid), cysteine (thiol, reduced or oxidized di-thiol), glutamine
(ethylcathoxyamide),
glutamic acid (propanoic acid), glycine (H), histidine (methyleneimidazole),
isoleucine (1-
methylpropane), leucine (2-methylpropane), lysine (butyleneamine), methionine
(ethylmethylthioether), phenylalanine (benzyl), proline (R3 forms a cyclic
ring with Ra and
the adjacent nitrogen group to form a pyrrolidine group), hydroxyproline,
serine (methanol),
threonine (ethanol, 1-hydroxyethane), tryptophan (methyleneindole), tyrosine
(methylene
phenol) or valine (isopropyl);

CA 02818308 2013-05-16
38
m (within this context) is an integer from 1 to 100,1 to 75,1 to 60,1 to 55,1
to 50,1 to 45,1
to 40,2 to 35,3 to 30,1 to 15,1 to 10,1 to 8,1 to 6,1,2,3,4 or 5;
n (within this context) is an integer from about 1 to 100, about 1 to 75,
about 1 to 60, about 1
to 50, about 1 to 45, about 1 to 35, about 1 to 25, about 1 to 20, about 1 to
15,2 to 10, about 4
to 12, about 5 to 10, about 4 to 6, about 1 to 8, about 1 to 6 , about 1 to 5,
about 1 to 4, about
1 to 3, etc.) or
Another linker according to the present invention comprises a polyethylene
glycol
linker containing from 1 to 1 to 100,1 to 75,1 to 60,1 to 55,1 to 50,1 to 45,1
to 40,2 to 35,
3 to 30,1 to 15,1 to 10,1 to 8,1 to 6,1,2,3,4 or 5 ethylene glycol units, to
which is bonded
a lysine group (preferably at its carboxylic acid moiety) which binds one or
two DNP groups
to the lysine at the amino group(s) of lysine. Still other linkers comprise
amino acid residues
(D or L) to which are bonded to ABT moieties, in particular, DNP, among others
at various
places on amino acid residue as otherwise described herein. In another
embodiment, as
otherwise described herein, the amino acid has anywhere from 1-15 methylene
groups
separating the amino group from the acid group in providing a linker to the
ABT moiety.
Or another linker is according to the chemical formula:
Where Z and Z' are each independently a bond, -(CH2)1-0, -(C112)1-S, -(CH2)1-N-
R ,
R2 R2 R2
0
= -(CH2); R2
or -Y-C-Y-
wherein said -(CH2); group, if present in Z or Z', is bonded to a connector,
ABT or CBT;
Each R is H, or a C1-C3 alkyl or alkanol group;
Each R2 is independently H or a C1-C3 alkyl group;
Each Y is independently a bond, 0, S or N-R;

CA 02818308 2013-05-16
39
Each i is independently 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45,
1 to 40,2 to 35, 3
to 30, 1 to 15, 1 to 10,1 to 8, 1 to 6, 1, 2, 3, 4 or 5;
D is
0
-(CHDrif- .
_______________ (CHAT-XI
Or
a bond, with the proviso that Z, Z' and D are not each simultaneously bonds;
j is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35,
3 to 30, 1 to 15, 1 to
10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5;
m' is Ito 100,1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3
to 30, 1 to 15, 1 to
10,1 to 8, 1 to 6, 1, 2, 3, 4 or 5;
n is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35,
3 to 30, 1 to 15, 1 to
10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5;
XI is 0, S or N-R; and
R is as described above, or a pharmaceutical salt thereof.
The term "connector", symbolized in the generic formulas by [CT], is used to
describe a chemical moiety which is optionally included in bifunctional
compounds
according to the present invention which forms from the reaction product of an
activated
ABT-linker with a PTB moiety (which also is preferably activated) or an ABT
moiety with
an activated linker-PTB as otherwise described herein. The connector group is
often the
resulting moiety which forms from the facile condensation of two or more
separate chemical
fragments which contain reactive groups which can provide connector groups as
otherwise
described to produce bifunctional or multifunctional compounds according to
the present
invention. It is noted that a connector may be distinguishable from a linker
in that the
connector is the result of a specific chemistry which is used to provide
bifunctional
compounds according to the present invention wherein the reaction product of
these groups
results in an identifiable connector group or part of a connector group which
is

CA 02818308 2013-05-16
distinguishable from the linker group, although in certain instances,
incorporated into the
linker group, as otherwise described herein. It is noted also that a connector
group may be
linked to a number of linkers to provide multifunctionality (i.e., more than
one PBT moiety
and/or more than one ABT moiety within the same molecule. It is noted that
there may be
some overlap between the description of the connector group and the linker
group such that
the connector group is actually incorporated or forms part of the linker,
especially with
respect to more common connector groups such as amide groups, oxygen (ether),
sulfur
(thioether) or amine linkages, urea or carbonate ¨0C(0)0- groups as otherwise
described
herein. It is further noted that a connector (or linker) may be connected to
ABT, a linker or
PBT at positions which are represented as being linked to another group using
the using the
symbol cijS Where two or more such groups are present in a linker or
connector, any of
an ABT, a linker or a PBT may be bonded to such a group. Where that symbol is
not used,
the link may be at one or more positions of a moiety.
Common connector groups which are used in the present invention include the
following chemical groups:
0
0
A N
NNN
A
1NA 1¨X2NA
0 0 0
1-X3X3k sAS
Nr1
or
Where X2 is 0, S, NR4, S(0), S(0)2, -S(0)20, -OS(0)2, or OS(0)20;
X3 is 0, S, NR4; and
R4 is H, a C1-C3 alkyl or alkanol group, or a -C(0)(C1-C3) group. The triazole
group,
indicated above, is a preferred connector group.

CA 02818308 2013-05-16
41
As discussed hereinabove, it is noted that each of the above groups may be
further
linked to a chemical moiety which bonds two or more of the above connector
groups into a
multifunctional connector, thus providing complex multifunctional compounds
comprising
more than one ABT and/or PBT group within the multifunctional compound.
Initial work by the inventors involved in identifying bifunctional compounds
ARM-
HI according to the present invention, began with the small molecule BMS-
378806 (Formula
1, below), (4-benzoy1-1-(2-(4-methoxy-1H-pyrrolo(2,3-b) pyridin-3-y1)-1,2-
dioxoethyl)-2-
methyl-, (2R)- Piperazine, CAS Number 357263-13-9, MW 406), a known inhibitor
of the
CD4-gp120 interaction. (Wang, et al. J. Med. Chem. 2003, 46, 4236-42396).
It was shown in international application PCT/US2010/52344 (published as
WO/2011/046946), which is incorporated by reference herein, that it was
possible to
derivatize Formula 1, at the carbon atom of the C4 methoxy group, in which the
carbon atom
of the C4 methoxy group could be replaced with various bulky substituents,
(Wang, J, S.; Le,
N.; Heredia, A .: Song, H. J.: Redfield, R.: Wang, L. X. Org. Biomol. Chem.
2005, 3, 1781-
1786) so as to provide a linker which would attract DNP without sacrificing
the compound's
ability to inhibit viral entry. This hypothesis was supported by an analysis
of a published
computational docking model suggesting that the C4 methoxy group in Formula 1
points
toward the solvent in the complex. See Kong. R.; Tan, J.; Ma, X.; Chen, W.;
Wang. C:.
Biochim. Biophys. Acta 2006, 1764. 766-7728.
4 Me
11
o N NO2
pc(A-14y) '''ttt'-' *(-'0:1:'"' ION
MN Me 2 No2
BMS378806 (1)
Na+1 N 2
HN N\%/1 "454µ143. ION
r"N N tr- \
0 0 0 4
(ARM-H)
3
Thus, in accordance with the invention of PCT/US2010/52344 (published as
WO/2011/046946), Formula 1 was re-engineered to include the capability to
recruit anti-DNP
antibodies to gp120-expressing particles (infected cells or viruses),
increasing the "visibility"

CA 02818308 2013-05-16
42
of the combination to the human immune system. Consequently, an ARM-HI of
Formula 4
was prepared in high yield (38% overall) via azide-alkyne cycloaddition
(Rostovtsev, V. V.;
Green, L. G.; Fokin, V. V.; Sharpless, K. B. Angew. Chem., lnt, Ed. 2002, 41,
2596-2599.
Tornoe, C.; Christensen, C.; Meldal, M. .J. Org. Chem. 2002, 67, 3057-3064) of
the
compounds of Formula 2 and Formula 3, which were derived in turn from known
intermediates. See, Wang, T.; et al. J. Med. Chem. 2003,46, 4236-4239.
As discussed above, ARM-HI compounds, including compounds according to the
present invention, target HIV by inhibiting virus entry while targeting Env-
expressing cells
for immune recognition and clearance. (See Fig. 1) Compounds set forth in the
prior PCT
application were shown to inhibit CD4 binding to HIV-1 gp120 and to out-
compete the CD4-
gp120 interaction. It was confirmed that ARM-HI has the ability to recruit
antibodies to
gp120 both in vitro and in tissue culture. Initial ELISA experiments
demonstrated a
concentration-dependent increase in anti-DNP antibody binding to the ARM-HI-
gp120
complex but not to gp120 alone. Thus, ARM-HI is capable of templating a
ternary complex
that also includes gp120 and anti-DNP antibody.
It was also confirmed that the ternary association could form in a complex
cellular
milieu, and that ARM-H bifunctional compounds have the ability to recruit anti-
DNP
antibodies to HIV-Env-expressing Chinese hamster ovary cells (CHO-gp120cells).
Thus, the
previous results presented in PCT/US2010/52344 (published as WO/2011/046946)
provide
strong evidence that ARM-HI bifunctional agents of the present invention are
capable of
recruiting anti-hapten (e.g. anti-DNP) antibodies to cells expressing the Env
glycoprotein in a
fashion that depends upon its simultaneous binding to both gp120 and anti-DNP
antibodies
and that the ternary complex formed from anti-DNP antibody, ARM-HI, and alive
Env-
expressing cell activates complement proteins and mediates cellular death.
Notably, in the absence of anti-DNP antibody and complement-preserved serum
(data
in green), in cells lacking the Env glycoprotein(CHO-WT, data in black), or in
the presence
of compound which lacks the DNP group, no cell death is observed, suggesting
that
termolecular complex formation is necessary for complement-dependent
cytotoxicity (CDC)
and that ARM-HI itself is not toxic to cells.

CA 02818308 2013-05-16
43
The present invention takes a novel approach and is directed to the
development of
further novel compositions which recruit anti-DNP antibodies and other anti-
hapten
antibodies, endogenous in most patients, to HIV via binding to the gp120
envelope protein,
which additionally prevents HIV from binding to CD4 and T4 cells, providing
novel
compositions and therapy for treating HIV infection and the symptoms
associated therewith.
The present compounds exhibit substantially greater activity than the
compounds which are
disclosed in PCT/US2010/52344 (published as WO/2011/046946).
The following detailed description outlines the design and synthesis of a
number of
bifunctional small-molecules capable of redirecting endogenous anti-hapten
antibodies,
especially including anti-dinitrophenyl (DNP) antibodies selectively to HIV,
and inducing
antibody-directed, cell-mediated cytotoxicity, which are based upon the
results obtained for
the compounds originally presented in PCT/US2010/52344 (WO/2011/046946).
The following chemical synthesis which is presented in Scheme 1 (Figure 3) and
Scheme 2 (Figure 4) may be used to synthesize the compound labeled as C-5
Furan in Figure
6 which shows exceptional activity as an anti-HIV agent. The Scheme 1 and
Scheme 2
chemical syntheses are genericized in Scheme 3, Figure 5, to provide generic
methods (either
directly or by analogy) for producing virtually all of the compounds which are
described
herein.
By way of synthesis, the carboxylic acid azide compound 2 (the azide readily
forming
a triazole connector molecule with an acetylenic group which links the ABT
group with the
PBT group) is prepared as otherwise described herein. Pursuant to scheme 1,
Figure 3, the
oligo(ethylene oxide) azide compound 1 is modified in sodium hydride and
solvent (THF)
with bromoacetic acid to provide compound 2, which contains both an
electrophilic moiety
(the carboxy group can condense to form an amide with an amine group) and an
azide which
can react with an acetylenic group to form a triazole (connector group).
The pathogen binding terminus (PBT) group in the present application is
modified to
contain an aryl group (ARYL2) on carbocyclic group of the indole bicyclic
ring. Scheme 2
(Figure 4) provides a rather facile synthesis of C-5 furan from the bromo-
substituted indole
compound 4, which condenses a substituted furan compound (4) onto the indole
ring as
indicated in Scheme 2 to produce the furan-substituted PTB compound 5.
Compound 5 is

CA 02818308 2013-05-16
44
treated with trifluoroacetic acid in dichloromethane to produce intermediate 6
which is
reacted with carboxyl azide compound 2 to produce compound 7. Compound 7 is
then
reacted with a compound containing an acetylenic moiety and an ABT group (in
Scheme 2, a
DNP group) under favorable conditions to produce the active bifunctional
compound C5-
furan (containing a PBT group containing a furanyl group and an ABT group
linked together
through a linking group, see Figure 6).
The chemical synthesis provided above may be presented in a more generalized
fashion as set forth in Scheme 3, Figure 5. In the generic synthesis a bromo-
substituted
indole compound is first reacted with oxalyl chloride followed by protected
piperazine in
trifluoroacetic acid to produce compound 10. An aryl group substituting for
the bromo group
in the indole moiety may be introduced as Arene 1 (Scheme 3, Figure 5) by
reacting an aryl
hydroxy boron substituted compound containing an alkylene group substituted
with a
hydroxyl, an amine or a sulfhydryl group which provides compound 11, which may
be
further reacted with carboxylic acid azide compound 2 to form an azide
containing compound
which is then further reacted with an acetylene containing group (containing a
linker group
and a ABT group (DNP) to condense the acetylenic group onto the azide to form
a triazole
containing compound 13. Compound 13 may be further reacted with an
appropriately
substituted (carboxylic acid group which can be condensed onto free amine
group of the
piperazine moiety) aryl group (arene 2, Figure 5) to form the final bioactive
biofunctional
compounds according to the present invention. As noted, this generic synthesis
may be used
to provide a larger number of compounds which can accommodate numerous aryl
groups and
numerous ABT groups as indicated. Various analogs are also synthesized,
specifically
exemplary compounds of the invention which include alternative ABT
substituents.
Combing an ABT group (with or without a further linking group) containing an
acetylenic
group with an azide is rather facile and the formation of an azide group
and/or an acetylenic
group may be used to generally link the ABT group to the PBT group through a
connector/linker as otherwise described herein.
Thusly, in the present invention a PBT portion of a molecule is derivatized
with a
linker containing an azide group which can form a connector molecule in
subsequent
reactions. Once the derivative PBT molecule is formed, bifunctional compounds
according
to the present invention may be formed by condensation with appropriate ABT-
containing
molecules to produce the final bifunctional compounds according to the present
invention.

CA 02818308 2013-05-16
Using the above synthesis with appropriate modification, bifunctional
compounds
according to the present invention may be readily synthesized. These compounds
contain a
single PBT moiety to which is linked a compound comprising an ABT moiety.
The above schemes provide exemplary synthesis of compounds according to the
present invention with various iterations of same provided by analogy using
well known
methods as described herein and as understood by those of ordinary skill in
the art. It is
noted that the experimental section provides significant detail to allow the
facile synthesis of
a variety of bifunctional compounds as otherwise described herein. The schemes
are not to
be considered limiting in setting forth teachings which provide compounds
according to the
present invention.
Turning to the biological data of bifunctional compounds according to the
present
invention, with reference to Fig. 6, this figure shows a number of compounds
which were
tested in a viral inhibition assay. In this assay, IC50's of a number of prior
art compounds
were determined against different HIV-1 isolates as set forth in Table 1
below. In this assay,
viral inhibition was determined by HIV Tat-induced luciferase (Luc) reporter
gene expression
after a single round of virus infection in TZM-bl cells according to the
method of Platt, et al.,
1998, 72, 2855-64. This biological data evidences that the present compounds
are
unexpectedly more active than are the compounds having a linker and ABT moiety
at
different positions of the indole ring, an unexpected result.
Compound JR-FL ADA HXBc2 SF162 BaL
Figure 6
BMS-378806 2595 344 31.9 359 dnc
ARM-hi 290 1003 dnc Dnc dnc
BMS-furan 0.117 0.152 0.097 0.005 0.01
m-phenyl 52.8 375 208 20800 150257
* Note- All
C5-furan 0.720 6.35 10.3 309 34.8
(Present Invention) values in nM;
dnc =does not converge

CA 02818308 2013-05-16
46
While specific analogs have been shown and described, the present invention is
not
limited to these specific analogs and other antibody recruiting compounds that
can function
as the antibody recruiting terminus connected by a linker to a binding
terminus that will bind
to the HIV glycoprotein gp120 (gp120 on the viral membrane as well as gp120
displayed on
infected cells), would fall within the scope of the present invention. All of
these compounds
can be formulated into pharmaceutical compositions as otherwise described
herein and used
in the methods which are presented.
Pharmaceutical compositions comprising combinations of an effective amount of
at
least one bifunctional compound according to the present invention, and one or
more of the
compounds otherwise described herein, all in effective amounts, in combination
with a
pharmaceutically effective amount of a carrier, additive or excipient,
represents a further
aspect of the present invention.
The compositions of the present invention may be formulated in a conventional
manner using one or more pharmaceutically acceptable carriers and may also be
administered
in controlled-release formulations. Pharmaceutically acceptable carriers that
may be used in
these pharmaceutical compositions include, but are not limited to, ion
exchangers, alumina,
aluminum stearate, lecithin, serum proteins, such as human serum albumin,
buffer substances
such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride
mixtures of
saturated vegetable fatty acids, water, salts or electrolytes, such as
prolamine sulfate,
disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride,
zinc salts,
colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-
based substances,
polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes,
polyethylene-
polyoxypropylene-block polymers, polyethylene glycol and wool fat.
The compositions of the present invention may be administered orally,
parenterally,
by inhalation spray, topically, rectally, nasally, buccally, vaginally or via
an implanted
reservoir. The term "parenteral" as used herein includes subcutaneous,
intravenous,
intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal,
intrahepatic,
intralesional and intracranial injection or infusion techniques. Preferably,
the compositions
are administered orally, intraperitoneally or intravenously.

CA 02818308 2013-05-16
47
Sterile injectable forms of the compositions of this invention may be aqueous
or
oleaginous suspension. These suspensions may be formulated according to
techniques
known in the art using suitable dispersing or wetting agents and suspending
agents. The
sterile injectable preparation may also be a sterile injectable solution or
suspension in a non-
toxic parenterally-acceptable diluent or solvent, for example as a solution in
1, 3-butanediol.
Among the acceptable vehicles and solvents that may be employed are water,
Ringer's
solution and isotonic sodium chloride solution. In addition, sterile, fixed
oils are
conventionally employed as a solvent or suspending medium. For this purpose,
any bland
fixed oil may be employed including synthetic mono- or di-glycerides. Fatty
acids, such as
oleic acid and its glyceride derivatives are useful in the preparation of
injectables, as are
natural pharmaceutically-acceptable oils, such as olive oil or castor oil,
especially in their
polyoxyethylated versions. These oil solutions or suspensions may also contain
a long-chain
alcohol diluent or dispersant, such as Ph. Hely or similar alcohol.
The pharmaceutical compositions of this invention may be orally administered
in any
orally acceptable dosage form including, but not limited to, capsules,
tablets, aqueous
suspensions or solutions. In the case of tablets for oral use, carriers which
are commonly used
include lactose and corn starch. Lubricating agents, such as magnesium
stearate, are also
typically added. For oral administration in a capsule form, useful diluents
include lactose and
dried corn starch. When aqueous suspensions are required for oral use, the
active ingredient is
combined with emulsifying and suspending agents. If desired, certain
sweetening, flavoring
or coloring agents may also be added.
Alternatively, the pharmaceutical compositions of this invention may be
administered
in the form of suppositories for rectal administration. These can be prepared
by mixing the
agent with a suitable non-irritating excipient which is solid at room
temperature but liquid at
rectal temperature and therefore will melt in the rectum to release the drug.
Such materials
include cocoa butter, beeswax and polyethylene glycols.
The pharmaceutical compositions of this invention may also be administered
topically. Suitable topical formulations are readily prepared for each of
these areas or organs.
Topical application for the lower intestinal tract can be effected in a rectal
suppository
formulation (see above) or in a suitable enema formulation. Topically-
acceptable

CA 02818308 2013-05-16
48
transdermal patches may also be used.
For topical applications, the pharmaceutical compositions may be formulated in
a
suitable ointment containing the active component suspended or dissolved in
one or more
carriers. Carriers for topical administration of the compounds of this
invention include, but
are not limited to, mineral oil, liquid petrolatum, white petrolatum,
propylene glycol,
polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. In
certain
preferred aspects of the invention, the topical cream or lotion may be used
prophylatically to
prevent infection when applied topically in areas prone toward virus
infection. In additional
aspects, the compounds according to the present invention may be coated onto
the inner
surface of a condom and utilized to reduce the likelihood of infection during
sexual activity.
Alternatively, the pharmaceutical compositions can be formulated in a suitable
lotion
or cream containing the active components suspended or dissolved in one or
more
pharmaceutically acceptable carriers. Suitable carriers include, but are not
limited to, mineral
oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl
alcohol, 2-
octyldodecanol, benzyl alcohol and water.
For ophthalmic use, the pharmaceutical compositions may be formulated as
micronized suspensions in isotonic, pH adjusted sterile saline, or,
preferably, as solutions in
isotonic, pH adjusted sterile saline, either with our without a preservative
such as
benzylalkonium chloride. Alternatively, for ophthalmic uses, the
pharmaceutical
compositions may be formulated in an ointment such as petrolatum.
The pharmaceutical compositions of this invention may also be administered by
nasal
aerosol or inhalation. Such compositions are prepared according to techniques
well-known in
the art of pharmaceutical formulation and may be prepared as solutions in
saline, employing
benzyl alcohol or other suitable preservatives, absorption promoters to
enhance
bioavailability, fluorocarbons, and/or other conventional solubilizing or
dispersing agents.
The amount of compound in a pharmaceutical composition of the instant
invention
that may be combined with the carrier materials to produce a single dosage
form will vary
depending upon the host and disease treated, the particular mode of
administration.
Preferably, the compositions should be formulated to contain between about
0.05 milligram

CA 02818308 2013-05-16
49
to about 750 milligrams or more, more preferably about 1 milligram to about
600 milligrams,
and even more preferably about 10 milligrams to about 500 milligrams of active
ingredient,
alone or in combination with at least one other bifunctional compound
according to the
present invention or other anti-HIV agent which may be used to treat HIV
infection or a
secondary effect or condition thereof.
It should also be understood that a specific dosage and treatment regimen for
any
particular patient will depend upon a variety of factors, including the
activity of the specific
compound employed, the age, body weight, general health, sex, diet, time of
administration,
rate of excretion, drug combination, and the judgment of the treating
physician and the
severity of the particular disease or condition being treated.
A patient or subject (e.g. a male human) suffering from HIV infection can be
treated
by administering to the patient (subject) an effective amount of the ARM-HI
compound
according to the present invention including pharmaceutically acceptable
salts, solvates or
polymorphs, thereof optionally in a pharmaceutically acceptable carrier or
diluent, either
alone, or in combination with other known antiviral or pharmaceutical agents,
preferably
agents which can assist in treating HIV infection, including AIDS or
ameliorate the
secondary effects and conditions associated with HIV infection. This treatment
can also be
administered in conjunction with other conventional HIV therapies.
These compounds can be administered by any appropriate route, for example,
orally,
parenterally, intravenously, intradermally, subcutaneously, or topically, in
liquid, cream, gel,
or solid form, or by aerosol form.
The active compound is included in the pharmaceutically acceptable carrier or
diluent
in an amount sufficient to deliver to a patient a therapeutically effective
amount for the
desired indication, without causing serious toxic effects in the patient
treated. A preferred
dose of the active compound for all of the herein-mentioned conditions is in
the range from
about 10 ng/kg to 300 mg/kg, preferably 0.1 to 100 mg/kg per day, more
generally 0.5 to
about 25 mg per kilogram body weight of the recipient/patient per day. A
typical topical
dosage will range from 0.01-5% wt/wt in a suitable carrier.

CA 02818308 2013-05-16
The compound is conveniently administered in any suitable unit dosage form,
including but not limited to one containing less than lmg, 1 mg to 3000 mg,
preferably 5 to
500 mg of active ingredient per unit dosage form. An oral dosage of about 25-
250 mg is
often convenient.
The active ingredient is preferably administered to achieve peak plasma
concentrations of the active compound of about 0.00001-30 mM, preferably about
0.1-30
M. This may be achieved, for example, by the intravenous injection of a
solution or
formulation of the active ingredient, optionally in saline, or an aqueous
medium or
administered as a bolus of the active ingredient. Oral administration is also
appropriate to
generate effective plasma concentrations of active agent.
The concentration of active compound in the drug composition will depend on
absorption, distribution, inactivation, and excretion rates of the drug as
well as other factors
known to those of skill in the art. It is to be noted that dosage values will
also vary with the
severity of the condition to be alleviated. It is to be further understood
that for any particular
subject, specific dosage regimens should be adjusted over time according to
the individual
need and the professional judgment of the person administering or supervising
the
administration of the compositions, and that the concentration ranges set
forth herein are
exemplary only and are not intended to limit the scope or practice of the
claimed
composition. The active ingredient may be administered at once, or may be
divided into a
number of smaller doses to be administered at varying intervals of time.
Oral compositions will generally include an inert diluent or an edible
carrier. They
may be enclosed in gelatin capsules or compressed into tablets. For the
purpose of oral
therapeutic administration, the active compound or its prodrug derivative can
be incorporated
with excipients and used in the form of tablets, troches, or capsules.
Pharmaceutically
compatible binding agents, and/or adjuvant materials can be included as part
of the
composition.
The tablets, pills, capsules, troches and the like can contain any of the
following
ingredients, or compounds of a similar nature: a binder such as
microcrystalline cellulose,
gum tragacanth or gelatin; an excipient such as starch or lactose, a
dispersing agent such as
alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate
or Sterotes; a

CA 02818308 2013-05-16
51
glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose
or saccharin; or a
flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
When the dosage
unit form is a capsule, it can contain, in addition to material of the above
type, a liquid carrier
such as a fatty oil. In addition, dosage unit forms can contain various other
materials which
modify the physical form of the dosage unit, for example, coatings of sugar,
shellac, or
enteric agents.
The active compound or pharmaceutically acceptable salt thereof can be
administered
as a component of an elixir, suspension, syrup, wafer, chewing gum or the
like. A syrup may
contain, in addition to the active compounds, sucrose as a sweetening agent
and certain
preservatives, dyes and colorings and flavors.
The active compound or pharmaceutically acceptable salts thereof can also be
mixed
with other active materials that do not impair the desired action, or with
materials that
supplement the desired action, such as other anti-HIV agents, antibiotics,
antifungals, anti-
inflammatories, or antiviral compounds. In certain preferred aspects of the
invention, one or
more ARM-HI compounds according to the present invention are coadministered
with
another anti-HIV agent and/or another bioactive agent, as otherwise described
herein.
Solutions or suspensions used for parenteral, intradermal, subcutaneous, or
topical
application can include the following components: a sterile diluent such as
water for
injection, saline solution, fixed oils, polyethylene glycols, glycerine,
propylene glycol or
other synthetic solvents; antibacterial agents such as benzyl alcohol or
methyl parabens;
antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such
as
ethylenediaminetetraacetic acid; buffers such as acetates, citrates or
phosphates and agents
for the adjustment of tonicity such as sodium chloride or dextrose. The
parental preparation
can be enclosed in ampoules, disposable syringes or multiple dose vials made
of glass or
plastic.
If administered intravenously, preferred carriers are physiological saline or
phosphate
buffered saline (PBS).
In one embodiment, the active compounds are prepared with carriers that will
protect
the compound against rapid elimination from the body, such as a controlled
release

CA 02818308 2013-05-16
52
formulation, including implants and microencapsulated delivery systems.
Biodegradable,
biocompatible polymers can be used, such as ethylene vinyl acetate,
polyanhydrides,
polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for
preparation of
such formulations will be apparent to those skilled in the art.
Liposomal suspensions may also be pharmaceutically acceptable carriers. These
may
be prepared according to methods known to those skilled in the art, for
example, as described
in U.S. Pat. No. 4,522,811 (which is incorporated herein by reference in its
entirety). For
example, liposome formulations may be prepared by dissolving appropriate
lipid(s) (such as
stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl
phosphatidyl
choline, and cholesterol) in an inorganic solvent that is then evaporated,
leaving behind a thin
film of dried lipid on the surface of the container. An aqueous solution of
the active
compound are then introduced into the container. The container is then swirled
by hand to
free lipid material from the sides of the container and to disperse lipid
aggregates, thereby
forming the liposomal suspension.
Detailed Synthetic Information
Materials and General Information: Purchased starting materials were used as
received
unless otherwise noted. All moisture sensitive reactions were performed in an
inert, dry
atmosphere of nitrogen in flame dried glassware. Reagent grade solvents were
used for
extractions and flash chromatography. Reaction progress was checked by
analytical thin-
layer chromatography (TLC, Merck silica gel 60 F-254 plates). The plates were
monitored
either with UV illumination, or by charring with anisaldehyde (2.5% p-
anisaldehyde, 1%
AcOH, 3.5% 112SO4(conc.) in 95% Et0H) or ninhydrin (0.3% ninhydrin (w/v), 97:3
Et0H-
AcOH) stains. Flash column chromatography was performed using silica gel (230-
400
mesh). The solvent compositions reported for all chromatographic separations
are on a
volume/volume (v/v) basis. ELISA and CDC experiments were performed in
triplicate and
repeated at least three times unless otherwise noted. I
Instrumentation: 1H-NMR spectra were recorded at either 400 or 500 MHz and are
reported
in parts per million (ppm) on the 6 scale relative to CDC13 (6 7.26) as an
internal standard
unless otherwise noted. Data are reported as follows: chemical shift,
multiplicity (s =
singlet, d =doublet, t =triplet, q =quartet, br =broad, m =multiplet),
coupling constants

CA 02818308 2013-05-16
53
(Hz), and integration. 13C-NMR spectra were recorded at either 100 or 125 MHz
and are
reported in parts per million (ppm) on the 8 scale relative to CDC13 (8
77.00). High
resolution mass spectra (HRMS) were recorded on a 9.4T Bruker Qe FT-ICR MS
(W.M.
Keck Facility, Yale University). Analytical ultra high-performance liquid
chromatography-
mass spectrometry (UPLC/MS) was performed on a Waters UPLC/MS instrument
equipped
with a reverse-phase C18 column (1.7 gm particle size, 2.1 x 50 mm), dual
atmospheric
pressure chemical ionization (API)/electrospray (ESI) mass spectrometry
detector, and
photodiode array detector. Samples were eluted with a linear gradient of 20%
acetonitrile-
water¨>100% acetonitrile containing 0.1% formic acid over 3 min at a flow rate
of 0.8
mL/min. Analytical UPLC/MS data are represented as follows: m/z; retention
time (Rt) in
minutes. High Pressure Liquid Chromatography (HPLC) using a Dynamax Rainin
Solvent
Delivery System equipped with a Varian Prostar Detector (Galaxie
Chromatography Data
System version 1.8.505.5), and absorbance measurements were made at 214 and
254 nm
simultaneously. A Waters Xterra Prep MS C18 7.8x150mm column was used for semi-
preparative purifications using a watenacetonitrile (A:B) gradient containing
0.1% TFA at
5.0 mL/min, as specified below for individual compounds. Analytical HPLC
analysis was
performed using a Varian C8 4.6x250mm Microsorb C8 column run at a flow rate
of 1.0
mL/min water:acetonitrile (A:B) gradient containing 0.1% TFA. Unless otherwise
noted, all
micro-plate based assays were quantitated using a BioTek Synergy 3 Microplate
reader and
data was fitted and graphed using GraphPad Prism version 5.00 for Windows
(GraphPad
Software, San Diego California USA, www.graphpad.com) or KaleidaGraph (Synergy
Software).
HO\/0\)1( N3 bromoaceti NaH/THF c
=
HOO \,)(N3
0
1 acid
0 2
Scheme 1. Synthesis of 2.
Azido polyethylene glycol 6 (0.60 g, 2 mmol, 1 equiv.) was dissolved in
H 11G-1-cr-t3N5 dry THF (10 mL) and cooled at 0 C, then sodium hydride (0.15
g, 6.3
mmol, 3.1 equiv.) was added in portions followed by bromoacetic acid (0.35 g,
2.5 mmol,

CA 02818308 2013-05-16
54
1,25 equiv.). The suspension was stirred at room temperature under nitrogen
overnight. Water
(1 mL) was added carefully and then stirred for 5 min. The reaction mixture
was concentrated
in vacuo. Dichloromethane was added and organic layer was washed with 2N HC1
and brine.
The organic layer was dried over Na2SO4 and all solvents were evaporated. Pure
6 (0.72 g,
98%) was obtained as an oil. 1H NIVIR (400 MHz, CDC13) 5 4.16 (s, 2H), 3.76
(s, 2H), 3.72
¨ 3.58 (m, 20H), 3.39 (s, 2H). MS (ES+) 366 [M +H]; 338 [M +H - N2]
O& 0trVIC)
f
(-fa Hiõ,tatia",w, di 0 -1 TFAiDCM 011NJ
Wora,"1
lihi 0
Or 117*(5-11118oc e
. 1FA
Gos
6
8
0
Oble 0 N)110
cuaik. Wm, pis)
=
2 MW
Of
EDC, Hag, DIPEA H
ia:12
a =
80%
IrtaN:13 aft
ftlYN; ife
NOI
Scheme 2. Synthesis of 9 from known 3.
/10:1 To a microwave vial (2.0 -5.0 mL) containing 3 (52 mg, 0.11 mmol) in
0 DMF/water (3.0/1.8mL), added 5-((B0C-Amino)methyl)furan-2-
boronic acid (Combi-Blocks LLC, San Diego CA; 37 mg, 0.154 mmol,
*Moo
1.4 equiv) and NaHCO3 (12.8 mg, 0.154 mmol, 1.4 equiv). Oxygen was
removed from the solvent by bubbling nitrogen gas in solution for 10 min, to
which
Pd(PPh3)4 (6.3 mg, 0.0055 mmol, 5 mol %) was added. The subsequent
heterogenous
solution was capped and heated in microwave reactor for 12 mm at 150 C When
LC/MS
analysis showed reaction completion. The volatile solvents were removed by
rotary
evaporation and crude material was purified by flash chromatography (flash
chromatography
(CombiFlash Automated Chromatographer, 12g colum; gradient elution ranging
from 0%

CA 02818308 2013-05-16
methanol:dichloromethane to 15% methanol:dichloromethane was performed over 30
column
volumes) to yield pure 5 as a yellow solid (52 mg, 0.088 mmol, 80%). 111 NMR
(500 MHz,
CDC13) 8 1126 (s, 111), 8.11 (d, J =2.6, 1H), 7.40 (bs, 511), 6.69 (d, J =8.4,
111), 6.52 (d, J =
3.2, 1H), 6.25 (d, J =3.2, 111), 5.20 (t, J =6.2, 1H), 4.30 (d, J = 6.4, 2H),
3.93 (s, 1H), 3.91 ¨
3.33 (m, 8H), 1.45 (s, 9H). UPLC/MS: (ES+) m/z (M 41)1- ; Rt =
r,pritti To 5 (51 mg, 0.86 mmol) in dichloromethane (800 L), added
4
trifluoroacetic acid (250 lit), resulting in a color change from yellow to
* e dark
brown. Solution was stirred at room temperature, open to air for 1
. WA
hr when TLC (20:1 dichloromethane/methanol) showed reaction
completion. Volatiles were removed by rotary evaporation, co-evaporating
several times
with chloroform, resulting in 6 as a yellow solid which was used without
further purification.
111 NMR (400 MHz, CDC13) 8 7.99 (s, 1H), 7.51 ¨7.33 (m, 611), 6.64 (d, J =
8.3, 1H), 6.54
(d, J =3.1, 1H), 6.27 (s, 1H), 3.90 (s, 3H), 3.46 (s, 8H).
To 2 (350 mg, 0.959 mmol 1.3 equiv)) dissolved in dichloromethane
* (20 mL), added diisopropylethylamine (250 !IL), EDC HC1 (300
mg,
1.56 mmol), HOBt (250 mg, 1.63 mmol) followed by 6 (350 mg,
r1.0"9:3 0.719 mmol). After 5 hrs, TLC (20:1 dichloromethane:methanol)
showed reaction completion. Reaction mixture was diluted with dichloromethane
(10 mL)
and washed with saturated NaHCO3 (3 x 30 mL), 2 M HC1 (1 x 30 mL) and brine (1
x 30
mL) and subsequently dried over anhydrous Mg504. Solution was filtered and
volatile
solvents were removed by rotary evaporation. Crude yellow-orange product was
purified by
flash chromatography (flash chromatography (CombiFlash Automated
Chromatographer, 24g
colum; gradient elution ranging from 0% methanol:dichloromethane to 10%
methanol:dichloromethane was performed over 30 column volumes) to yield pure 7
as a light
yellow sticky solid (360 mg, 0.088 mmol, 60%). 'H NMR (400 MHz, CDC13) 8 11.32
(s,
1H), 8.12 (d, J =3.3, 1H), 8.07 (s, 1H), 7.38 (d, J =8.1,611), 6.67 (d, J
=8.4, 1H), 6.51 (d, J
=3.2, 1H), 6.29 (d, J =3.2, 1H), 4.52 (d, J =6.2, 211), 4.01 (s, 211), 3.92
(s, 3H), 3.80-3.42
(m, 28H), 3.32 ¨ 3.25 (m, 2H).
a To a
microwave vial containing 7 (200 mg, 0240 mmol)
NONA. in tBuOH/water (2 mL/2 mL), added alkyne 8 (100 mg,
41111
Lza
!PM
4 LILPEPt

CA 02818308 2013-05-16
56
0.261 mmol, 1.1 equiv), followed by 120 L of 0.1M CuSO4 and 240 IAL of 0.1M
sodium
ascorbate. The reaction mixture was capped and heated to 130 C for 30 minutes
in
microwave reactor when LC/MS showed reaction completion. Volatile solvents
were
removed by rotary evaporation and crude product was purified by HPLC (30-50%
B, 36 min;
Rt =27.03 min). Like fractions were combined and volatile solvents were
removed by rotary
evaporation, resulting in 9 as a yellow sticky solid (281 mg, 0.228 mmol, 95
%). 111 NMR
(400 MHz, CDC13) 6 11.21 (s, 1H), 9.09 (d, J =2.5, 1H), 8.77 (s, 111), 8.18
(m, 3H), 7.74 (s,
1H), 7.52¨ 7.34 (m, 6H), 6.92 (d, J = 9.5, 1H), 6.69 (d, J =8.4, 1H), 6.54 (d,
J =3.2, 1H),
6.32 (d, J =3.1, 111), 4.65 (s, 2H), 4.58 (d, J =6.0, 2H), 4.45 (t, J =4.9,
2H), 4.08 (s, 2H),
3.94 (s, 3H), 3.83 ¨ 3.28 (m, 46H). HRMS (ES+) calc'd for C581-174N10020 (M
4H) m/z
1231.5154. Found 12315178; for (M+Na)+, calc'd 1253.4973, found 1253.5045.
Anal.
HPLC Retention Time =27.89 min at 0-60% B, 36 min
1) NaHCO3
Pd(PF113)4, MW
B(OH) 1) EDC, HOBT,
2
OW r-N-Fnix DIPEA
0
OMe 0 (-14-12 1102C10nN3 2
TEA I ¨ wax
=
2) Fmoc-C6u 0 2)10% pip
Br 3) TFA / 0 13 3) CuS0 , NaAsc,
1.420/10H,
Ni-I2
NEUF 8
n'
0
1101Me 0 r-N1-1 NO2
OMe r Nj ARLMNE
"u2 Add 0 NO2
0 EDC, HOBT, =
/ 0 IrN.,,,40/-=,)/n.
DIPEA \,N-J NO2
16 NH-Ir-ko/ n
0
0
Scheme 3. Representative general route to additional analogs
To a microwave vial (2.0 -5.0 mL) containing 10 (200 mg, 0.546 mmol, available
from
Patent No. WO 2011046946) in DMF/water (3.0/2mL), added 5-
OMe 0 NH
((BOC-Amino)methyl)furan-2-boronic acid (Combi-Blocks LLC,
40o San Diego CA; 241 mg, 0.622 mmol, 1.14 equiv) and NaHCO3 (50
o mg, 0.595 mmol, 1.1 equiv). Oxygen was removed from the
N HBoc

CA 02818308 2013-05-16
57
solvent by bubbling nitrogen gas in solution for 10 mm, to which Pd(PPh3)4 (30
mg, 0.026
mmol, 5 mol %) was added. The subsequent heterogenous solution was capped and
heated in
microwave reactor for 15 mm at 150 C when LC/MS analysis showed reaction
completion.
The volatile solvents were removed by rotary evaporation and crude material
was purified by
flash chromatography (CombiFlash Automated Chromatographer, 24g colum;
gradient
elution ranging from 0% methanol:dichloromethane to 15%
methanol:dichloromethane was
performed over 30 column volumes) to yield pure 11 as a brown solid (225 mg,
0.467 mmol,
85%). 1H NMR (400 MHz, CDC13) 8 11.23 (s, 1H), 8.09 (s, 1H), 7.33 (d, J =8.2,
1H), 6.63
(d, J =8.2, 111), 6.48 (d, J =3.2, 1H), 6.23 (d, J =3.2, 111), 5.36 (s, 1H),
4.29 (s, 211), 3.90
(brs, 511), 3.64 (brs, 2H), 3.18 (brs, 2H), 3.09 (brs, 211), 1.43 (s, 9H).
OMe 0 r
NFmoc To 11(400 mg, 0.83 mmol) dissolved in dichloromethane (20
el I 0 mL), added
Fmoc-OSU (560 mg, 1.66 mmol, 2 equiv) followed
by diisopropylethylamine (500 4). Resulting yellow-orange
z 0
mixture was stirred at room temperature under an atmosphere of
NHBoc
nitrogen for 12hr when TLC (10:1 dichloromethane-methanol)
showed reaction completion. The organic mixture was washed with a saturated
solution of
ammonium chloride (3 x 20 mL) and brine (1 x 30 mL) and then dried over
anhydrous
magnesium sulfate. All volatiles were removed by rotary evaporation and crude
material was
purified by flash chromatography (CombiFlash Automated Chromatographer, 24g
colum;
flushing with dichloromethane for 5 column volumes then gradient elution
ranging from 0%
methanol:dichloromethane to 5% methanol:dichloromethane was performed over 20
column
volumes) to yield pure 12 as a yellow solid (400 mg, 0.567 mmol, 69%). 1H NMR
(400
MHz, CDC13) 11.25 (s, 111), 8.12 (d, J =3.1, 111), 7.76 (brs, 2H), 7.55 (brs,
2H), 7.42 (m,
3H),7.31 (brs, 2H), 6.70 (d, J =8.3, 1H), 6.54 (d, J =3.1, 1H), 6.26 (d, J
=3.1, 1H),5.11 (m,
111), 4.52 (d, J =6.2, 211), 4.32 (d, J =6.4, 2H), 4.24 (brs, 1H), 3.94 (s,
311), 3.42 (m, 811),
1.47 (s, 911).
OMe 0 (" NFmoc
1µ1µ) To 12
dissolved (600 mg, 0.85 mmol) in dichloromethane (15
I 0 mL),
carefully added trifluoracetic acid (6 mL) and let stir at
0 room
temperature under an atmosphere of nitrogen when TLC
NH2 (10:1
dichloromethane-methanol) indicated reaction completion

CA 02818308 2013-05-16
58
(2 hr). All volatiles were removed by rotary evaporation and crude green
residue was
azeotroped with chloroform (3 x 10 mL) and used without further purification,
yielding 13 as
a TFA salt as brown solid (570 mg, 96%). III NMR (400 MHz, CDC13) 8 11.25 (s,
1H),
9.85 (s, 3H), 8.28 (s, 2H), 7.94 (s, 111), 7.70 (s, 211), 7.48 (s, 211), 7.34
(s, 211), 7.16 (d, J =
7.5, 1H), 6.52 (d, J =8.1, 1H), 6.46 (s, 1H), 6.40 (s, 111), 4.46 (d, J =4.4,
2H), 4.20 (s, 211),
3.82 (s, 3H), 3.61 ¨ 3.11 (m, 8H).
OMe 0 NFmoc
To a solution of 13 (300 mg, 0.510 mmol) in dichloromethane
N
0 (25 mL), added 2
(200 mg, 0.722 mmol, 1.4 equiv), added EDC-
N
HC1 (180 mg, 3 equiv), HOBT (150 mg, 3 equiv) and
o
NN y-k0
diisopropylethylamine (180 L) and let stir under an atmosphere
of nitrogen until TLC (20:1 dichloromethane-methanol) indicated
reaction completion (5 hrs). The organic mixture was washed with saturated
ammonium
chloride (1 x 30 mL) and saturated sodium bicarbonate (1 x 30 mL) and
volatiles were
removed in vacuo. Resulting crude material was purified by flash
chromatography
(CombiFlash Automated Chromatographer, 24g colum; flushing with
dichloromethane for 5
column volumes then gradient elution ranging from 0% methanol:dichloromethane
to 5%
methanol:dichloromethane was performed over 20 column volumes) to yield pure
14 as a
yellow residue (380 mg, 0.44mmol, 86%). III NMR (400 MHz, CDC13) 8 11.27 (s,
1H), 8.12
(d, J =3.2, 111), 7.91 (t, J =6.1, 1H), 7.75 (brs, 2H), 7.55 (brs, 211), 7.46-
7.26 (m, 5H), 6.69
(d, J =8.3, 111), 6.52 (d, J =3.2, 111), 6.30 (d, J =3.2, 111), 451 (m, 411),
4.23 (brs, 111), 4.04
(s, 211), 3.94 (s, 3I1), 3.75 ¨ 3.26 (m, 24H).
OMe 0 (--11H
To 14 (288mg, 0.33 mmol) in dichloromethane (2 mL), added
N
o piperadine (350 IAL) and let stir under an atmosphere of nitrogen at
N3 room
temperature until TLC (20:1 dichloromethane-methanol)
o rpi 4
NH -Irk() showed reaction
completion (2 hr). All volatiles were removed in
vacuo and crude material was purified flash chromatography
(CombiFlash Automated Chromatographer, 12g colum; gradient elution ranging
from 0%
methanol:dichloromethane to 20% methanol:dichloromethane was performed over 30
column
volumes) to yield pure 15 as a yellow residue (170 mg, 0.265 mmol, 80%). 1H
NMR (400
MHz, CDCb) 8 11.25 (s, 111), 8.10 (s, 111), 7.97 (s, 1H), 7.38 (d, J =8.3,
111), 6.67 (d, J =
8.4, 111), 6.50 (d, J =3.2, 111), 6.29 (d, J =3.2, 111), 4.50 (d, J =6.2,
2I1), 4.02 (s, 211), 3.94

CA 02818308 2013-05-16
59
(s, 3H), 3.83 ¨3.72 (m, 2H), 3.70 ¨ 3.46 (m, 141-1), 3.34 ¨ 3.25 (m, 2H), 3.17
¨ 3.05 (m, 2H),
3.05 ¨2.96 (m, 2H), 2.96 ¨ 2.82 (m, 2H).
To a microwave vial containing 15 (160 mg,
OMe 0 NOJH NO2
0.25 mmol) in water/tBuOH (2.3 mL/2.3 mL),
I
0 NI___41-374 40
No2 added alkyne 8, 0.1M CuSO4 (117 L) in water
V 0and 0.1 M sodium ascorbate (234 L). The
NH.irko
reaction mixture was capped and heated to 130
C for 30 minutes in microwave reactor when
LC/MS showed reaction completion. Volatile solvents were removed by rotary
evaporation
and crude product was purified by flash chromatography (CombiFlash Automated
Chromatographer, 12g colum; gradient elution ranging from 0%
methanol:dichloromethane
to 40% methanol:dichloromethane was performed over 140 column volumes) to
yield 16 as a
yellow residue (255 mg, 0.246 mmol, 98%). 1H NMR (400 MHz, CDC13) 8 1125 (s,
1H),
9.06 (d, J =2.5, 1H), 8.75 (brs, 111), 8.19 (d, J =9.5, 1H), 8.10 (s, 1H),
7.92 (brs, 1H), 7.66
(s, 11-1), 7.37 (d, J =8.3, 111), 6.89 (d, J =9.5, 111), 6.67 (d, J =8.3,
111), 6.51 (d, J =3.2, 1H),
6.28 (d, J =3.1, 1H), 4.62 (brs, 2H), 4.49 (d, J =6.1, 2H), 4.43 (t, J =5.0,
211), 4.04 (s, 3H),
3.95 (brs, 4H), 3.91-3.41 (m, 34H), 3.13 (m, 3H).
General Synthetic Procedure for coupling of aryl carboxylic acids to (16) ¨See
Table 1.
To a 16 (10 mg, 0.01 mmol) in dichloromethane (1 mL), added arene carboxylic
acid (0.018
mmol, 1.8 equiv), EDC-HC1 (3.5 mg, 0.018 mmol, 1.8 equiv), HOBT (3.0 mg, 0.019
mmol,
1.9 equiv) and diisopropylethylamine (10 AL). Resulting solution was allowed
to stir at room
temperature under an atmosphere of nitrogen until LC/MS indicated reaction
completion (5-
14 hrs). Resulting solution was diluted with dichloromethane (5 mL) and then
washed with a
saturated solution of sodium bicarbonate (5 mL) and ammonium chloride (5 mL).
The
organic layer was dried over anhydrous magnesium sulfate, filtered and
volatiles were
removed in vacuo. Crude material was purified by HPLC.
Coupling Product (18). 1H NMR (400 MHz, CDC13) 8 11.28 (s, 1H), 9.07 (s, 1H),
8.76 (s,
111), 8.20 (d, J =9.4, 111), 8.11 (s, 111), 7.97 (s, I H), 7.67 (brs, 111),
7.43 ¨7.30 (m, 21-1), 7.22
¨ 7.09 (m, 3H), 6.90 (d, J = 9.5, 1H), 6.76 ¨ 6.63 (m, 111), 6.52 (brs, 1H),
6.29 (brs, 111), 4.63
(s, 2H), 4.47 (m, 41-1), 4.05 (s, 31-1), 3.96 ¨ 3.33 (m, 4011), 2.32 (d, J
=11.8, 3H) (Note: peak
broadening effect of configurational isomerism about benzoyl-piperazine amide
bond).

CA 02818308 2013-05-16
Coupling Product (19).1H NMR (400 MHz, CDC13) 6 11.20 (s, 111), 9.06 (d, J
=2.6,111),
8.74 (s, 1H), 8.20 (d, J =6.9, 1H), 8.12 (s, 1H), 8.00 (s, 114), 7.67 (s, 1H),
7.39 (d, J =8.3,
1H), 7.33 ¨ 7.27 (m, 2H), 7.21 ¨ 7.13 (m, 1H), 6.89 (d, J =9.5, 1H), 6.68 (d,
J =8.3, 111),
6.52 (d, J =3.2, 111), 6.30 (d, J =3.2, 1H), 4.63 (s, 2H), 4.50 (m, 211), 4.43
(m, 211), 4.07 (s,
2H), 3.95 (s, 3H), 3.78 ¨ 3.42 (m, 38H), 2.38 (s, 3H).
Coupling Product (20).1H NMR (400 MHz, CDC13) 8 11.25 (s, 111), 9.07 (d, J
=2.5, 111),
8.75 (s, 11-1), 8.20 (dd, J =2.3, 9.4, 111), 8.12 (s, 1H), 7.99 (s, 1H), 7.67
(s, 1H), 7.39 (d, J =
8.3, 1H), 7.32 (m, 114), 7.23 (m, 2H), 6.90 (d, J =9.5, 1H), 6.68 (d, J =8.4,
1H), 6.52 (d, J =
3.1, 1H), 6.29 (d, J =3.0, 1H), 4.62 (s, 2H), 4.49 (d, J =5.9, 211), 4.44 (m,
211), 4.06 (s, 211),
3.95 (s, 314), 3.89-3.49 (m, 38H), 2.38 (s, 3H).
Coupling Product (21).1H NMR (400 MHz, CDC13) 8 11.21 (s, 1H), 9.05 (d, J
=2.5, 1H),
8.74 (s, 1H), 8.19 (d, J =9.5, 1H), 8.11 (m, 211), 7.63 (s, 1H), 7.36 (m,
411), 7.24 ¨ 7.18 (m,
111), 6.88 (d, J =9.5, 1H), 6.68 (d, J =8.3, 114), 6.52 (d, J =3.1, 1H), 6.30
(d, J =3.1, 1H),
4.59 (s, 2H), 4.52 ¨ 4.24 (m, 8H), 4.08 (s, 2H), 3.94 (s, 3H), 3.85 ¨ 3.33 (m,
3414), 2.68 (m,
211), 124 (t, J =7.4, 3H).
Coupling Product (22).1H NMR (400 MHz, CDC13) 8 11.14 (s, 1H), 9.05 (d, J
=2.5,111),
8.74 (s, 1H), 8.24 ¨ 8.09 (m, 3H), 7.67 (s, 111), 7.37 (m, 311), 7.29 (brs,
111), 6.88 (d, J =9.5,
111), 6.69 (d, J =8.4, 1H), 6.53 (d, J =3.1, 1H), 6.31 (d, J =3.1, 111), 4.60
(s, 211), 4.50 (d, J
=5.8, 211), 4.41 (d, J =4.7, 211), 4.10 (brs, 2H), 3.94 (s, 3H), 3.77-3.42 (m,
3811), 3.04 ¨2.79
(m, 1H), 1.25 (d, J =6.5, 6H).
Coupling Product (23). 1H NMR (500 MHz, CDC13) 6 11.08 (s, 1H), 9.05 (d, J
=2.6, 1H),
8.73 (s, 111), 8.19 (dd, J =2.6, 9.5, 1H), 8.13 (brs, 211), 7.92 (m, 4H), 7.71
(s, 1H), 757 (s,
214), 7.49 (d, J =8.1, 111), 7.40 (d, J =8.3, 1H), 6.88 (d, J =9.5, 111), 6.70
(d, J =8.1, 111),
6.54 (d, J =3.1, 1H), 6.33 (d, J =3.2, 111), 4.63 (s, 211), 4.52 (s, 2H), 4.42
(s, 2H), 4.11 (s,
211), 3.95 (s, 3H), 3.77 (d, J =5.1,411), 3.74-3.45 (m, 3411).
Coupling Product (24).1H NMR (500 MHz, CDCI3) 6 11.27 (s, 111), 9.07 (d, J
=2.7, 111),
8.75 (s, 1H), 8.20 (dd, J =2.6, 9.5, 1H), 8.12 (d, J =3.2, 1H), 7.91 (s, 1H),
7.66 (s, 111), 7.39
(d, J = 8.3, 1H), 7.37 ¨ 7.32 (m, 114), 7.24 (d, J =1.4,111), 7.02 (d,J =8.2,
1H), 6.88 (m, 2H),
6.69 (d, J =8.4, 1H), 6.52 (d, J =3.3, 111), 6.29 (d, J =3.2,111), 4.61 (s,
2H), 4.49 (d, J =6.1,
211), 4.43 (m, 211), 4.06 (s, 311), 3.95-3.50 (m, 4011), 2.01 (s, 111).

CA 02818308 2013-05-16
61
Coupling Product (25).1H NMR (500 MHz, CDC13) 5 11.22 (s, 1H), 9.06 (d, J
=2.6, 1H),
8.74 (s, 1H), 8.19 (d, J =9.5, 1H), 8.08 (d, J = 3.3, 1H), 8.03 ¨ 7.89 (brs,
1H), 7.70¨ 7.49 (m,
111), 7.39 (d, J =8.3, 1H), 7.25-7.2 (m, 111), 6.91-6.87 (m, 3H), 6.68 (d, J
=8.4, 1H), 6.53 (d,
J = 3.3, 111), 6.31 (d, J = 3.2, 11-1), 4.57 (s, 211), 4.49 (d, J = 6.2, 2H),
4.45 ¨ 4.25 (m, 21-1),
4.07 (s, 2H), 3.94 (s, 3H), 3.83 ¨3.39 (m, 38H), 2.01 (s, 1H).
Coupling Product (26).1H NMR (400 MHz, CDC13) 5 11.17 (s, 1H), 9.03 (d, J
=2.4, 1H),
8.73 (s, 111), 8.17 (d, J =9.5, 111), 8.09 (brs, 211), 7.63 (s, 1H), 7.38 (d,
J = 8.3, 1H), 7.31 ¨
7.27 (m, 111), 6.88-6.82 (m, 311), 6.68 (d, J =8.3, 1H), 6.52 (d, J =2.9, 1H),
6.31 (d, J =2.8,
1H), 4.57 (s, 2H), 4.49 (d, J =4.9, 211), 4.44-4.22 (m, 2H), 4.08 (s, 211),
3.93 (s, 311), 3.83 ¨
3.43 (m, 38H).
Coupling Product (27). 1H NMR (400 MHz, CDC13) 11.18 (s, 111), 8.97 (d, J
=2.3, 1H),
8.69 (s, 1H), 8.12 (d, J =9.5, 1H), 8.00 (m, 211), 7.64 (brs, 111), 7.33 (d, J
=7.6, 111), 6.83 (d,
J =9.6, 1H), 6.62 (d, J =8.0, 1H), 6.50 (brs, 1H), 6.47-6.31 (m, 311), 6.28
(brs, 1H), 4.57
(brs, 2H), 4.48 (brs, 2H), 4.43 ¨ 4.31 (m, 2H), 4.04 (brs, 211), 3.88 (s, 3H),
3.79 ¨ 3.35 (m,
38H).
Coupling Product (28). 1H NMR (400 MHz, CDC13) 5 11.23 (s, 1H), 9.06 (s, 111),
8.75 (s,
111), 8.19 (d, J =9.5, 111), 8.11 (brs, 211), 7.60 (s, 111), 7.40 (m, 5H),
6.89 (d, J =9.5, 1H),
6.68 (d, J =8.4, 111), 6.52 (s, 111), 6.30 (s, 111), 4.74 (s, 2H), 4.57 (s,
2H), 4.49 (s, 2H), 4.40
(s, 2H), 4.09 (s, 2H), 3.95 (s, 3H), 3.86 ¨ 3.30 (m, 38H), 1.25 (s, 1H).
Coupling Product (29).1H NMR (400 MHz, CDC13) 5 11.23 (s, 1H), 9.06 (s, 1H),
8.75 (s,
1H), 8.70 ¨ 8.49 (m, IH), 8.20 (d, J = 9.2, 111), 8.13 (brs, 1H), 8.03 (brs,
1H), 7.88 (brs, 111),
7.71 (brs, 111), 7.63 (brs, 111), 7.38 (m, 211), 6.89 (d, J =9.5, 111), 6.68
(brs, 1H), 6.52 (brs,
1H), 6.30 (brs, 1H), 4.59 (brs, 211), 4.48 (brs, 2H), 4.41 (brs, 211), 4.08
(brs, 2H), 3.95 (s,
3H), 3.92¨ 3.12 (m, 3811), 2.00 (s, 311).
Coupling Product (30). 1H NMR (400 MHz, CDC13) 5 11.28 (s, 1H), 9.03 (d, J
=2.6, 111),
8.73 (m, 311), 8.16 (m, 211), 8.08 (s, 111), 8.01 (m, 1H), 7.60 (brs, 1H),
7.50 (s, 111), 7.36 (d, J
=8.3, 111), 6.86 (d, J =9.5, 111), 6.67 (d, J =8.4, 111), 650 (d, J =3.3,
111), 6.28 (d, J =3.3,
1H), 4.53 ¨4.42 (m, 411), 4.35 (brs, 2H), 4.09 (brs, 211), 3.94 (s, 3H), 3.79
¨ 3.39 (m, 38H).

CA 02818308 2013-05-16
62
Coupling Product (31). 1H NMR (400 MHz, CDC13) 6 11.20 (s, 111), 9.01 (s, 1H),
8.90 (s,
211), 8.73 (s, 114), 8.17 (s, 2H), 8.09 (s, 1H), 7.70 (s, 2H), 7.54 (s, 111),
7.38 (s, 111), 6.86 (brs,
1H), 6.69 (brs, 1H), 6.52 (brs, 1H), 6.30 (brs, 1H), 4.50 (m, 411), 4.38 (brs,
2I1), 4.11 (brs,
211), 3.99 - 3.46 (m, 411-1).
e 18 19 20 21 22 23
410 * 10 10 * "Cc, 0110
Et
CD4 0-29 5.14 1.36 0.447 0.469 0.953 524
1
MT-2 0.01 -25 -0.6 0.025 -0.6 NIA -5.0
24 25 28 27 28 29 30 31
14Ciim (II
am
CD4 I 0.49 0.20 0.239 0.745 0.422 0.62 3.7 4.7
MT-2 02 0_09 0.09 0.23 -1.0 0.048 1.3 9.3
Table 1. Various synthesized compounds through general method as described
from
corresponding commercially available arene carboxylic acid. All activities are
in p.M.
Summary
The present invention meets the strategic need for a new treatment for HIV
infection
by providing bifunctional small molecules generally referred to as ARM-HI's
which function
through orthogonal pathways - both by inhibition the gp120-CD4 interaction,
and by
recruiting anti-DNP antibodies to gp120-expressing cells - in preventing the
cell infection
and spread of HIV. It is shown that: ARM-HI's according to the present
invention exhibit
substantially greater activity than ARM-H compounds previously published.
The present antiviral approach has distinct advantages over other small-
molecule,
protein, and vaccine-based anti-HIV strategies.

CA 02818308 2013-05-16
63
Although the human immune response has been demonstrated to generate
neutralizing
anti-gp120 antibodies around which the virus does not effectively mutate,
vaccine-based
approaches toward inducing such antibodies in human hosts have not yet proven
successful.
In theory, although the HIV virus mutates extremely rapidly in human hosts,
since it must
retain CD4-binding activity in order to remain infectious, antibody-recruiting
small
molecules that mimic the CD4 recognition motif such as the ARM-HI's of the
invention have
the hope of serving the same functional role as neutralizing anti-gp120
antibodies.
Furthermore, as small molecules, these materials likely possess substantial
advantages over
protein-based therapeutics including low propensity for immunogenicity, high
metabolic
stability, ready large-scale production, and relatively low cost.
The evidence suggests that a cellular immune response is necessary for viral
inactivation in vivo, and the bifunctional small molecules of the invention
have been shown
to directly target gp120-expressing particles to macrophages and neutophils.
This approach to antiviral therapy is also ideal as a prophylactic, as the
bifunctional
compound are not be expected to have any significant adverse side effects,
being only active
when virus is present.
The complete disclosure of all patents, patent applications, and publications,
and
electronically available material (including, for instance, nucleotide
sequence submissions in,
e.g., GenBank and RefSeq, and amino acid sequence submissions in, e.g.,
SwissProt, PIR,
PRF, PDB, and translations from annotated coding regions in GenBank and
RefSeq) cited
herein are incorporated by reference. Any inconsistency between the material
incorporated
by reference and the material set for in the specification as originally filed
shall be resolved in
favor of the specification as originally filed. The foregoing detailed
description and examples
have been given for clarity of understanding only. No unnecessary limitations
are to be
understood therefrom. The invention is not limited to the exact details shown
and described,
for variations obvious to one skilled in the art will be included within the
invention defined
by the following claims.
All headings are for the convenience of the reader and should not be used to
limit the
meaning of the text that follows the heading, unless so specified.

Representative Drawing

Sorry, the representative drawing for patent document number 2818308 was not found.

Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Application Not Reinstated by Deadline 2019-06-13
Inactive: Dead - No reply to s.30(2) Rules requisition 2019-06-13
Deemed Abandoned - Failure to Respond to Maintenance Fee Notice 2018-11-19
Inactive: Abandoned - No reply to s.30(2) Rules requisition 2018-06-13
Inactive: Office letter 2018-02-19
Inactive: Agents merged 2018-02-19
Revocation of Agent Request 2017-12-29
Appointment of Agent Request 2017-12-29
Inactive: S.30(2) Rules - Examiner requisition 2017-12-13
Inactive: Report - No QC 2017-12-08
Inactive: Office letter 2017-01-09
Revocation of Agent Requirements Determined Compliant 2017-01-09
Appointment of Agent Requirements Determined Compliant 2017-01-09
Inactive: Office letter 2017-01-09
Appointment of Agent Request 2016-12-02
Revocation of Agent Request 2016-12-02
Inactive: Adhoc Request Documented 2016-11-28
Letter Sent 2016-11-22
Request for Examination Requirements Determined Compliant 2016-11-16
All Requirements for Examination Determined Compliant 2016-11-16
Request for Examination Received 2016-11-16
Revocation of Agent Request 2016-11-03
Appointment of Agent Request 2016-11-03
Letter Sent 2015-02-12
Inactive: Single transfer 2015-01-29
Change of Address or Method of Correspondence Request Received 2015-01-29
Inactive: Cover page published 2013-08-09
Inactive: First IPC assigned 2013-06-20
Inactive: Notice - National entry - No RFE 2013-06-20
Inactive: IPC assigned 2013-06-20
Inactive: IPC assigned 2013-06-20
Inactive: IPC assigned 2013-06-20
Application Received - PCT 2013-06-20
National Entry Requirements Determined Compliant 2013-05-16
Application Published (Open to Public Inspection) 2012-05-24

Abandonment History

Abandonment Date Reason Reinstatement Date
2018-11-19

Maintenance Fee

The last payment was received on 2017-11-16

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Fee History

Fee Type Anniversary Year Due Date Paid Date
MF (application, 2nd anniv.) - standard 02 2013-11-18 2013-05-16
Basic national fee - standard 2013-05-16
MF (application, 3rd anniv.) - standard 03 2014-11-17 2014-10-29
Registration of a document 2015-01-29
MF (application, 4th anniv.) - standard 04 2015-11-17 2015-10-23
MF (application, 5th anniv.) - standard 05 2016-11-17 2016-10-24
Request for examination - standard 2016-11-16
MF (application, 6th anniv.) - standard 06 2017-11-17 2017-11-16
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
YALE UNIVERSITY
Past Owners on Record
CHRISTOPHER PARKER
DAVID SPIEGEL
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column (Temporarily unavailable). To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2013-05-15 63 2,846
Claims 2013-05-15 20 504
Abstract 2013-05-15 1 62
Drawings 2013-05-15 6 297
Cover Page 2013-08-08 1 40
Notice of National Entry 2013-06-19 1 195
Courtesy - Certificate of registration (related document(s)) 2015-02-11 1 125
Courtesy - Abandonment Letter (R30(2)) 2018-07-24 1 165
Reminder - Request for Examination 2016-07-18 1 117
Acknowledgement of Request for Examination 2016-11-21 1 175
Courtesy - Abandonment Letter (Maintenance Fee) 2018-12-30 1 174
PCT 2013-05-15 23 969
Correspondence 2015-01-28 2 49
Correspondence 2016-11-02 3 148
Request for examination 2016-11-15 1 51
Correspondence 2016-12-01 5 192
Courtesy - Office Letter 2017-01-08 4 155
Courtesy - Office Letter 2017-01-08 4 154
Examiner Requisition 2017-12-12 5 327
Courtesy - Office Letter 2018-02-18 1 34